LOGO

METABOLIC HEALTH



INTRODUCTION

I have developed the following information on Metabolic Health for my own reference and use, but it is available for anybody interested in Insulin Resistance / Diabetes etc.
I have included many scientific papers here and they mostly speak for themselves ie. I have made no value judgements or made any recommendations.

I am only concerned about Insulin Resistance, Pre-Diabetes and Type 2 Diabetes, meaning Metabolic Health
I am not a Doctor and do not warrant or assume any legal liability or responsibility for their accuracy, completeness, or usefulness. If you have health concerns see a Doctor.
I apologise for the fact that this study is highly plagerized and thus sort of disjointed as I switch from one source to another. I have provided many good sources below and some that can be problematic as they are considering just one aspect of the diabetes conundrum.


LINE1

Insulin resistance is a major feature of Metabolic Health. Insulin resistance is associated with Hyperglycemia, obesity, hypertension, dyslipidemia, nonalcoholic fatty liver disease, obstructive sleep apnea and sleep depravation, and some forms of cancer. This cluster of maladies has been termed "Insulin Resistance Syndrome." Therefore, an individual with insulin resistance is strongly predisposed to an increased risk of life-threatening clinical conditions, including cardiovascular disease.

TED1B

The model above depicts that with the development of insulin resistance the beta cells in the pancreas compensates by increasing the insulin response over time in years, thereby avoiding the development of measurable fasting hyperglycemia until the time that the increase in hyperglycemia (Impaired Fasting Glucose) overwhelms the insulin response or there is a decline in insulin secretion.


LINE1


TED2

The clinical consequences of insulin resistance are not due to insulin resistance per se but come from the hyperinsulinemia that occurs as the individual with insulin resistance attempts to maintain normoglycemia, Meaning compensatory hyperinsulinemia. Chronic hyperinsulinemia may be beneficial to resistant tissues requiring it, for example to maintain insulin action in liver, muscle, and adipose tissues; however, it may wreak havoc with tissues that have normal sensitivity to insulin. Even within the same tissue, some of the insulin-regulated pathways, such as the glucose metabolic pathway, are more resistant to insulin than others. Therefore, intensive efforts are being directed toward identifying novel nutritional and pharmacological approaches that improve insulin sensitivity in target tissues


The Pathophysiology of insulin resistance in type 2 diabetes mellitus Insulin resistance has an important role in the pathophysiology of T2DM and CVD and both genetic and environmental factors facilitate its development. More than 90% of people with T2DM are obese, and the release of free fatty acids (FFAs) and cytokines from adipose tissue directly impairs insulin sensitivity (Figure 6). In skeletal muscle and adipose tissue, FFA-induced reactive oxygen species (ROS) production blunts activation of insulin recep tor substrate 1 (IRS-1) and PI3K-Akt signalling, leading to down regulation of insulin responsive glucose transporter 4 (GLUT-4)



Insulin resistance is a complex condition influenced by multiple factors. The following are some of the key contributors:
1. Inflammation: Chronic inflammation can interfer with insulin signaling pathways, leading to insulin resistance. Conditions like obesity and autoimmune diseases often involve inflammation.
2. Stress: Prolonged stress increases the release of cortisol, a hormone that can promote insulin resistance by affecting glucose metabolism and increasing blood sugar levels.
3. Insulin: Paradoxically, high levels of insulin (hyperinsulinemia) can lead to insulin resistance. This occurs when the body produces more insulin to compensate for cells not responding effectively, eventually causing the cells to become even more resistant.
4. Urea: Elevated urea levels, often seen in kidney dysfunction, can impair insulin signaling and contribute to insulin resistance.
5. Seed Oils: Some studies suggest that excessive consumption of certain seed oils, high in omega-6 fatty acids, may promote inflammation and insulin resistance. However, this is still a topic of ongoing research.
6. Sleep Deprivation: Lack of sleep disrupts hormonal balance, including insulin and cortisol levels, leading to impaired glucose metabolism and increased insulin resistance.
7. Age: As we age, the risk of insulin resistance increases due to changes in body composition, decreased physical activity, and alterations in hormone levels2.
8. Obesity: Excess body fat, particularly around the abdomen, is a significant risk factor for insulin resistance. Fat cells release inflammatory cytokines that interfere with insulin signaling.
9. Sedentary Lifestyle: Physical inactivity reduces the body's ability to use insulin effectively. Regular exercise helps improve insulin sensitivity.
10. Diet: Diets high in refined carbohydrates, sugars, and unhealthy fats can contribute to insulin resistance. Conversely, a balanced diet rich in whole foods, fiber, and healthy fats can improve insulin sensitivity.
11. Genetics: Family history and genetic predisposition play a role in the development of insulin resistance. Certain genetic variations can affect how the body responds to insulin.
12. Hormonal Imbalances: Conditions like polycystic ovary syndrome (PCOS) and hormonal changes during menopause can increase the risk of insulin resistance.
13. Medications: Some medications, such as corticosteroids and certain antipsychotics, can induce insulin resistance as a side effect.

LINE1

T2D4


Type 2 Diabetes Mellitus (T2DM), one of the most common metabolic disorders, as shown above is caused by a combination of two primary factors: insufficient insulin secretion by pancreatic β-cells and/or the inability of insulin-sensitive tissues to respond appropriately to insulin (insulin resistance). Because insulin release and activity are essential processes for glucose homeostasis, the molecular mechanisms involved in the synthesis and release of insulin, as well as in its detection are tightly regulated. Defects in any of the mechanisms involved in these processes can lead to a metabolic imbalance responsible for the development of the disease.

Type 2 Diabetes Mellitus (T2DM) risk factors and the pathological changes leading to the perpetuation of insulin dysfunction. Complex combinations of genetic, metabolic and environmental factors that interact with one another constitute both non-modifiable (ethnicity and family history/genetic predisposition) and modifiable risk factors (obesity, low physical activity and an unhealthy diet). These states affect cell function resulting in a complex network of pathological changes that influence mutually and lead to the perpetuation of insulin dysfunction. ROS: reactive oxygen species; ER: endoplasmic reticulum; AGEs: advanced glycation end products; PKC: protein kinase C; LPS: lipopolysaccharide; miRNA: microRNA.
This will be the objective of the following:


LINE1

T2D5


The interplay of the pancreas with the brain, liver, gut as well as adipose and muscle tissue. The pancreas interacts with the brain, liver, gut and adipose and muscle tissue in a highly sophisticated network via various hormones, neurotransmitters and cytokines. BNDF, brain-derived neurotrophic factor; CCK, cholecystokinin; GIP, glucose-dependent insulinotropic peptide; GLP-1, glucagon-like peptide 1; GRP, gastrin-releasing peptide; IL-6, Interleukin 6; MCH, melanin concentrating hormone; NPY, neuropeptide Y; PACAP, pituitary adenylate cyclase-activating polypeptide; POMC, pro-opiomelanocortin; VIP, vasoactive intestinal peptide.


LINE1


In regards to most of the folowing informationn I am usually looking at the cell level and the biochemical pathways that make this study so interesting. I have also included some videos on cell mechanics at the bottom.

We stay alive because millions of different biochemical reactions are taking place inside our bodies all the time. Each of our cells is like the busy auto assembly line. Raw materials, half-finished products, and waste materials are constantly being used, produced, transported, and excreted. The "workers" on the cellular assembly line are mainly enzymes. These are the proteins that make biochemical reactions happen.

The sum of all the biochemical reactions in an organism is referred to as Metabolism. Metabolism includes both exothermic (Catabolic) chemical reactions and endothermic (Anabolic) chemical reactions.
Catabolic reactions break down molecules into smaller units and release energy. An example of a catabolic reaction is the breakdown of glucose during cellular respiration,(Glycolysis), which releases energy that cells need to carry out life processes.
Anabolic reactions absorb energy and build bigger molecules from smaller ones. An example of an anabolic reaction is the joining of amino acids to form a protein.( Protein Synthesis)

Metabolism is those life-sustaining chemical transformations within the cells of living organisms.
The three main purposes of metabolism are:
1. The conversion of food/fuel to energy (ATP) to run cellular processes,
2. The conversion of food/fuel to building blocks for proteins, lipids, nucleic acids, and some carbohydrates,
3. The elimination of Metabolic wastes.
4. Metabolism can be impacted by age, sex, DIET, exercise, sleep, and injury or disease.


The following are a few definitions and images of Cell processes.


cell2


A cell is the smallest living organism and the basic unit of life. 30 - 40 trillion cells make up the human body. Cells have three parts: the membrane, the nucleus, and the cytoplasm. A typical animal cell contains more than 42,000 different kinds of molecules. In the past 20 years, great progress has been made in understanding how these molecules combine and interact to form a living creature. In addition to this, there are approximately 40 trillion bacteria occuping the intestinal tract that need to be fed.
Essential Characteristics of Cells explained Here



Video: Biology of Cell Structure


Nucleus:
The nucleus represents the cell's headquarters. There is typically one nucleus per cell. However, this is not always the case. Skeletal muscle cells, for instance, have two.
The nucleus of the cell is a membrane-bound organelle found in most eukaryotic cells and is the largest organelle within the cell. It contains nearly all of the cell's genetic material, organized into chromosomes, which are essential for gene expression and cell function. The nuclear envelope, which surrounds the nucleus, is perforated with nuclear pores that regulate the transport of molecules between the nucleus and the cytoplasm. (refer here for more info.)



membrain1

Plasma Membrane:


To ensure each cell remains separate from its neighbor, a special membrane, known as the plasma membrane, envelops the cell. Phospholipids make most of this membrane and prevent water-based substances from entering the cell.
Note: Cholesterol plays a distinct role in membrane structure. Cholesterol will not form a membrane by itself, but inserts into a bilayer of phospholipids.
(refer here for more info.)





GLUCOSE AND INSULIN RECEPTORS

Cells need transporters for glucose and other water-based (hydrophilic) molecules because their cell membranes are made of a lipid bilayer that is hydrophobic (water-repelling) in nature. Imagine trying to mix oil and water—it just doesn’t work! Glucose and other hydrophilic molecules can't pass through the lipid bilayer on their own. To get these crucial molecules into the cell, specialized transport proteins act as gateways. These transporters provide a way for glucose and other water-soluble molecules to cross the cell membrane without getting stuck. It’s a bit like having a key to unlock the door and let in what the cell needs to function and thrive.

The plasma membrane contains a range of receptors, which carry out a number of tasks, including being:
1. Gatekeepers: Some receptors allow certain molecules through and stop others.
2. Markers: These receptors act as name badges, informing the immune system that they are part of the organism and not foreign invaders.
3. Communicators: Some receptors help the cell communicate with other cells and the environment.
4. Fasteners: Some receptors help bind the cell to its neighbors.

In total, there are hundreds of different types of receptors, each playing a crucial role in cellular communication and function.


GLUCOSE RECEPTORS

GLUT4-1 Refer to Source here

At this point in the discussion as we are concerned with insulin and insulin resistance and the two main receptors are the insulin and glucose receptors. There are approimately 14 glucose receptors and of those there are 5 of major concern.

An insulin-dependent glucose transporter, GLUT4, plays a crucial role in glucose regulation in Skeletal muscle, adipose cells (fat cells), the heart and some brain cells.
Muscle cells are significant consumers of glucose in the body. Approximately 80% of the glucose from food is processed by muscle cells. This high percentage underscores the importance of muscle tissue in maintaining overall metabolic health and regulating blood glucose levels.
GLUT4 is responsible for the facilitated diffusion of glucose into these cells. It is unique because its activity is regulated by insulin, making it essential for maintaining normal blood glucose levels.

GLUT4-4 Refer to Source here
Schematic of insulin-induced translocation of glucose transporter 4 from cytosol to the cell membrane. The binding of insulin to its receptors initiates a signal transduction cascade, which results in the activation of Akt. Akt acts on the glucose transporter 4 (GLUT4) containing vesicles in the cytosol to facilitate their fusion with the cell membrane. When more GLUT4 molecules are present in the membrane, the rate of glucose uptake is elevated. GLUT4: Glucose transporter 4.


LINE1

GLU19

Refer here for siurce information.

In order to ensure normal body function, the human body is dependent on a tight control of its blood glucose levels. Note the 4 to 6 mM is a Europe designation, that would approximate 70 to 100 mg/l in the us system as what would be normal fasting blood glucose levels. This arbitrary as the fasting level should be closer to the 80 range. a fasting Glucose level of 100 to 126 is considered pre diabetic and over 126 is Diabetic This balance is accomplished by a highly sophisticated network of various hormones and neuropeptides released mainly from the brain, pancreas, liver, intestine as well as adipose and muscle tissue. Within this network, the pancreas represents a key player by secreting the blood sugar-lowering hormone insulin and its opponent glucagon. However, disturbances in the interplay of the hormones and peptides involved may lead to metabolic disorders such as type 2 diabetes mellitus (T2DM) whose prevalence, comorbidities and medical costs take on a dramatic scale.
GLU19 above shows the Maintenance of blood glucose levels by glucagon and insulin. When blood glucose levels are low, the pancreas secretes glucagon, which increases endogenous blood glucose levels through glycogenolysis. After a meal, when exogenous blood glucose levels are high, insulin is released to trigger glucose uptake into insulin-dependent muscle and adipose tissues as well as to promote glycogenesis.


MECHANISM:
1. Insulin Binding: When blood glucose levels rise, insulin is released from the pancreas.
2. Signal Transduction: Insulin binds to its receptor on the cell surface, initiating a signaling cascade.
3. GLUT4 Translocation: This signaling causes GLUT4-containing vesicles within the cell to move to the plasma membrane.
4. Glucose Uptake: GLUT4 integrates into the membrane, allowing glucose to enter the cell from the bloodstream.

Importance
5. Energy Storage: In muscle cells, glucose is used for energy production or stored as glycogen.
6. Fat Storage: In adipose tissue, glucose is converted into fat for long-term energy storage.
7. Amino acids are converted to protein for muscle hypertrophy.
REGULATION:
8. Exercise: Physical activity can also stimulate GLUT4 translocation to the cell membrane, independent of insulin.
9. Insulin Resistance: In conditions like Type 2 Diabetes, the efficiency of GLUT4 translocation is impaired, leading to elevated blood glucose levels.
Understanding GLUT4's role helps in grasping how the body manages glucose and the impact of insulin resistance on metabolic health.


INSULIN RECEPTOR

This animation describes the role of the insulin receptor in type 2 diabetes. It focuses on the very recent discovery of how the hormone insulin actually binds to the receptor on the surface of cells, as determined by Professor Mike Lawrence's laboratory at the Walter and Eliza Hall Institute. Insulin binds to the receptor protein on the cell surface and instructs the cell to take up glucose from the blood for use as an energy source. In type 2 diabetes, they believe that insulin binds to the receptor normally, but the signal is not sent into the cell, the cells do not take up glucose and the resulting high blood glucose levels cause organ damage over time.


INSULIN9

PANCREAS
The pancreatic β-cell plays a key role in glucose homeostasis by secreting insulin, the only hormone capable of lowering the blood glucose concentration. Impaired insulin secretion results in the chronic hyperglycemia that characterizes type 2 diabetes (T2DM). The healthy β-cell acts as a glucose sensor matching its output to the circulating glucose concentration. It does so via metabolically induced changes in electrical activity, which culminate in an increase in the cytoplasmic Ca2+ concentration and initiation of Ca2+-dependent exocytosis of insulin-containing secretory granules.





CYTOPLASM

GLY2

The cytoplasm is the interior of the cell that surrounds the nucleus. It includes the organelles and a jelly-like fluid called the cytosol. Many of the important reactions that take place in the cell occur in the cytoplasm such as:
1. Glycolysis: This is the first stage of cellular respiration, where glucose is broken down into pyruvate, producing a small amount of ATP and NADH.
2. Protein Synthesis: The translation of mRNA into proteins occurs on ribosomes, which are often found in the cytoplasm.
3. Cell Division: Processes such as mitosis and meiosis, which are essential for cell replication and reproduction, begin in the cytoplasm.
3. Metabolic Pathways: Various metabolic pathways, including those involved in the synthesis and breakdown of molecules, occur in the cytoplasm.
These reactions are crucial for the cell's energy production, growth, and overall function and are discussed below.






Mitochondria:

MIT7 Refer to Source here


The mitochondrion is the organelle of a cell that is actively involved in the production of energy or fuel (ATP) for the basic biological functioning of a cell. ATP - or adenosine triphosphate - is a high-energy compound that fuels cellular energy requirements. It is produced by the process of cellular respiration that takes place in the mitochondrion. During cellular respiration the food is oxidized, oxygen is consumed, and carbon dioxide is released.
Cellular respiration is the metabolic process that occurs in the cells of living organisms. It involves converting chemical energy from nutrients, such as glucose, into adenosine triphosphate (ATP), which cells use for energy. This process also produces carbon dioxide and water as waste products.

The main stages of cellular respiration include:

IN THE CYTOPLASM:
Glycolysis: The breakdown of glucose into pyruvate, producing a small amount of ATP and NADH.
IN THE MITOCHONDRIA:
Citric Acid Cycle (Krebs Cycle, TCA Cycle): The pyruvate is further broken down, generating more NADH and FADH2, and releasing carbon dioxide.
Oxidative Phosphorylation (OXPHOS):
The NADH and FADH2 produced in the previous stages donate electrons to the electron transport chain, driving the production of a large amount of ATP. This process is essential for providing the energy required for various cellular activities and maintaining life.




ATP1

The ATP-ADP cycle:
the ATP-ADP cycle is an essential process in cellular energy metabolism.
ATP (adenosine triphosphate) is the energy-carrying molecule in cells. ATP releases energy when one of its three phosphate molecules breaks free,(Phosphorylation) becoming ADP (adenosine diphosphate). ADP can be recycled back into ATP by adding a phosphate molecule.
ATP is useful in many cell processes such as glycolysis, photosynthesis, beta oxidation, anaerobic respiration, active transport across cell membranes (as in the electron transport chain), and synthesis of macromolecules such as DNA.

Phosphorylation is an important process as the chemical addition of a phosphoryl group (PO3-) to an organic molecule. This process involves the transfer of a phosphate group from a donor molecule, typically ATP (adenosine triphosphate), to an acceptor molecule, such as a protein or small molecule.
1. Phosphorylation is a biochemical process that involves the addition of a phosphate group via an ester bond, usually catalyzed by enzymes called kinases.
2. It can occur on proteins, small molecules, or carbohydrates, leading to changes in their structure, function, or activity.
3. Phosphorylation is a reversible process, as enzymes called phosphatases can remove phosphate groups (dephosphorylation) from molecules.




EA1

An enzyme is a protein that speeds up a biochemical reactions. It is a biological catalyst. An enzyme generally works by reducing the amount of activation energy needed to start the reaction. the activation energy needed for glucose to combine with oxygen to produce carbon dioxide and water. The overall reaction releases energy, but an initial activation energy is needed to start the process. The activation energy without an enzyme is much higher than the activation energy when an enzyme is used.


Enzymes: principles and biotechnological applications


Key Metabolic Process Definitions

GLU18

1. Gluconeogenesis: Synthesis of glucose using non-carbohydrate precursors.
2. Glycolysis: Degradation of glucose into pyruvic acid and energy for cell metabolism.
3. Glycogenesis: Synthesis of glycogen from glucose.
4. Glycogenolysis: Degradation of glycogen into glucose.
5. Lipogenesis: Conversion of acetyl-CoA into fatty acids and subsequent triglyceride synthesis.
6. Lipolysis: Degradation of lipids and triglycerides into free fatty acids.
7. Protein synthesis: The biological process by which amino acids are assembled into specific polypeptides.
8. Apoptosis: A genetically directed process of cell self-destruction that is marked by the fragmentation of nuclear DNA.



WEHI Glycolysis

Glycolysis" by wehi.tv (2021)

Citric Acid Cycle (2020) by Drew Berry wehi.tv

Pyruvate Dehydrogenase Complex (2021) Drew Berry wehi.

Electron Transport Chain (2019) Drew Berry wehi.

Electron Transport Chain (2019) Drew Berry wehi.

Ninja video on Glycolysis



Pathophysiology of Type 2 Diabetes Mellitus

Regulatory principles in metabolism -Then and now

Diagnosis and Management of the Metabolic Syndrome

Cellular mechanisms of insulin resistance

Essentials of Cell Biology

Sympathetic nervous activation in metabolic syndrome and obesity

Insulin Resistance and Hyperinsulinemia

Metabolic syndrome: a sympathetic disease?

Saturated Fat with Dr. Ben Bikman

Insulin Resistance and Metabolic Syndrome with Dr. Ben Bikman

Albuminuria: An Underappreciated Risk Factor for Cardiovascular Disease

Enzymes: principles and biotechnological applications

Cell Metabolism

CORI CYCLE Gluconeogenesis

Glycolysis

Non-insulin dependent diabetes mellitus: the gathering storm

Social, clinical, and policy implications of ultra processed food addiction

2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes: Developed by the task force on the management of cardiovascular disease in patients with diabetes of the European Society of Cardiology

Insulin Secretion and Insulin Sensitivity Pattern Is Different in Isolated Impaired Glucose Tolerance and Impaired Fasting Glucose

Effect of physical activity and different exercise modalities on glycemic control in people with prediabetes: a systematic review and meta-analysis of randomized controlled trials

Distinct metabolic profile according to the shape of the oral glucose tolerance test curve is related to whole glucose excursion: a cross-sectional study

Insulin Resistance

Regulation of Cellular Respiration

Physiology, Aging

Protein acylation in the inhibition of insulin secretion by norepinephrine, somatostatin, galanin, and PGE2

The Disparate Effects of Epinephrine and Norepinephrine on Hyperglycemia in Cardiovascular Surgery

Association between triglyceride glucose index and biological aging in U.S. adults:

Defining and characterizing the progression of type 2 diabetes

From the Triumvirate to the Ominous Octet: A New Paradigm for the Treatment of Type 2 Diabetes Mellitus

ESC Guidelines on diabetes, pre-diabetes, and cardiovascular diseases

Improvement in Glycemic and Lipid Profiles in Type 2 Diabetics with a 90-Day Ketogenic Diet

CD38 ecto-enzyme in immune cells is induced during aging regulating NAD+ and NMN levels

NAD+ metabolism: pathophysiologic mechanisms and therapeutic potential


GLYCOLYSIS

GLY8

Glycolysis represents the process of breaking down monosaccharides, which involves the energy metabolism, homeostasis, and the linkage of various physiological functions such as muscle movement, development, neurotransmission, etc. what happens when there are Elevated levels of ATP and alanine create negative feedback to inhibit PK, does the flow reverese bak to g6p When there are elevated levels of ATP and alanine, they act as negative feedback inhibitors for pyruvate kinase (PK). This inhibition slows down the conversion of phosphoenolpyruvate (PEP) to pyruvate in glycolysis. As a result, the flow of metabolites is redirected upstream.

However, the flow does not reverse back to glucose-6-phosphate (G6P). Instead, the accumulation of PEP and other intermediates can lead to an increase in the levels of fructose-1,6-bisphosphate (F1,6BP), which can then be converted back to fructose-6-phosphate (F6P) and eventually to G6P through gluconeogenesis or other pathways

Phosphofructokinase is the main enzyme controlled in glycolysis. High levels of ATP, citrate, or a lower, more acidic pH decrease the enzyme’s activity. An increase in citrate concentration can occur because of a blockage in the citric acid cycle. Fermentation, with its production of organic acids like lactic acid, frequently accounts for the increased acidity in a cell; however, the products of fermentation do not typically accumulate in cells.

Biochemistry, Glycolysis

Glycolysis: A multifaceted metabolic pathway and signaling hub

Glycolysis

Glycolysis: A multifaceted metabolic pathway and signaling hub

Regulation of Glycolysis

Alcohol and Metabolic-associated Fatty Liver Disease

Aging and Insulin Resistance: Just Say iNOS

Regulation of Glycolysis: Short and Long-Term Control Mechanisms

WHPI Glycolysis


AGING

AGING

Metabolite and protein associations with general health in the population

Aging and Insulin Resistance: Just Say iNOS

Insulin Resistance Accelerates Biological Aging as Measured by Aging Clocks


IFG-IGT

IFG-IGT

Postchallenge Glucose, A1C, and Fasting Glucose as Predictors of Type 2 Diabetes and Cardiovascular Disease

Diagnosis, Prognosis, and Treatment of Impaired Glucose Tolerance and Impaired Fasting Glucose

Diabetic retinopathy

VIDEO Understanding Type 2 Diabetes

STATISTA: What share of adults are obese

Factors correlated with targeted prevention for prediabetes classified by impaired fasting glucose, impaired glucose tolerance, and elevated HbA1c:



GLUT4-3

GLU6

GLU13

GLY3

GLU9

GLU19

PAN16

AMPK1

PI3K5

BACK TO MENU




Insulin Resistance: From Mechanisms to Therapeutic Strategies

Assessment HOMA as a predictor for new onset diabetes mellitus and diabetic complications in non-diabetic adults

Insulin resistance - Reference pathway

What is Glycolysis and Why is it Important?


BACK TO MENU






METABOLIC SYNDROME
(FKA INSULIN RESISTANCE SYNDROME)

MS5

Metabolic syndrome represents the clinical diagnostic entity identifying those individuals at high risk with respect to the (cardiovascular) morbidity associated with insulin resistance
The National Cholesterol Education Program and other organizations have proposed that the MetS can be recognized clinically by a clustering of simple clinical measures including waist circumferences, blood pressure, triglycerides, high-density lipoproteins, and glucose. People with this clustering have most or all of the components of the MetS. Identifying the MetS has several advantages. It discovers persons who are at increased risk for cardiovascular disease. Increased age and metabolic syndrome are the most important relevant factors for diabetes mellitus, especially by using the International Diabetes Federation criteria for definition of the metabolic syndrome.

The Global Epidemic of the Metabolic Syndrome

Pathophysiology of Type 2 Diabetes Mellitus

The impact of diabetes on cognitive impairment and its progression to dementia

Overview of Metabolic Reactions

Chemical Reactions in Living Things

MS6

The Sympathetic‐Immune Milieu in Metabolic Health and Diseases: Insights from Pancreas, Liver, Intestine, and Adipose Tissues

Amino Acid Ingestion Strongly Enhances Insulin Secretion in Patients With Long-Term Type 2 Diabetes

Visceral adiposity index (VAI), lipid accumulation product (LAP), and product of triglycerides and glucose (TyG) to discriminate prediabetes and diabetes

Reassessment of Glucose Effectiveness and Insulin Sensitivity From Minimal Model Analysis

Diagnosis and Management of the Metabolic Syndrome

Lipid-Overloaded Enlarged Adipocytes Provoke Insulin Resistance Independent of Inflammation

Prognostic value of triglyceride glucose (TyG) index in patients with acute decompensated heart failure

Metabolic Inflexibility: When Mitochondrial Indecision Leads to Metabolic Gridlock

Metabolic Flexibility as an Adaptation to Energy Resources and Requirements in Health and Disease

Metabolic flexibility and insulin resistance

The association between advanced glycation end products (AGEs) and ABC (hemoglobin A1C, blood pressure, and low-density lipoprotein cholesterol) control parameters among patients with type 2 diabetes mellitus

Metabolic States of the Body

The Carbohydrate-Insulin Model of Obesity: Beyond ‘Calories In, Calories Out’

How monitoring ketones and glucose can help you achieve metabolic flexibility

Diagnosis and Management of the Metabolic Syndrome

Metabolic syndrome – a new definition and management guidelines

The Influence of Metabolic Syndrome inPredicting Mortality Risk Among US Adults: Importance of Metabolic Syndrome Even in Adults With Normal Weight

Regulation of insulin sensitivity by serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2

Non-insulin dependent diabetes mellitus: the gathering storm

2013 ESC Guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD

Metabolic Inflexibility: When Mitochondrial Indecision Leads to Metabolic Gridlock

Mechanisms of Insulin Action and Insulin Resistance

What’s on your table? How America’s diet has changed over the decades

Globally, calorie availability and the prominence of food in household spending are inversely related

Effect of circadian clock disruption on type 2 diabetes

Regulation of Glycolysis

Metabolism Disrupting Chemicals and Metabolic Disorders

Glycolysis Biochemistry

Glucose control of glucagon secretion

The Central Role of Glucokinase in Glucose Homeostasis

The somatostatin-secreting pancreatic δ-cell in health and disease

Monitoring and modelling the dynamics of the cellular glycolysis pathway

The role of glycolytic metabolic pathways in cardiovascular disease and potential therapeutic approaches

The Central Role of Glucokinase in Glucose Homeostasis

Metabolic Effects of Late Dinner in Healthy Volunteers

Video: What if Heart Disease and Diabetes had the same cause? | Ivor Cummins

The Metabolic Interplay between Cancer and Other Diseases

Molecular Mechanisms of Glucocorticoid-Induced Insulin Resistance

Molecular Mechanisms of Glucocorticoid-Induced Insulin Resistance

Assessing Insulin Sensitivity and Resistance in Humans

Circadian phase inversion causes insulin resistance in a rat model of night work and jet lag

Trapped fat: Obesity pathogenesis as an intrinsic disorder in metabolic fuel partitioning

Nonlinear dynamics of multi-omics profiles during human aging


Metabolic Syndrome, is a cluster of comorbid conditions including obesity, hypertension, and disordered carbohydrate and lipid metabolism, constitutes a significant health and social problems, which occur together more often than by chance alone:
Although obesity is a well-known risk factor for poor Metabolic Health, Metabolic Health issues such as insulin resistance and diabetes risk also affect normal-weight people. A useful method for assessing Metabolic health is to determine the presence of Metabolic Syndrome, which is defined as having 3 of the following 5 criteria: central obesity, elevated blood glucose, elevated triglycerides, low levels of high-density lipoprotein cholesterol, and elevated blood pressure.

Other than genetics, Metabolic Syndrome is highly correlated with unhealthy diet, unhealthy sleeping patterns, sedentary behaviors, and physical inactivity.

METABOLIC SYNDROME
Elevated waist circumference ≥ 40 inches in men;
Elevated waist circumference ≥ 35 inches in women;
Elevated triglycerides ≥150 mg/dL (1.7 mmol/L);
Reduced HDL-C ≤40 mg/dL (1.0 mmol/L) in males;
Reduced HDL-C ≤50 mg/dL (1.3 mmol/L) in females;
Elevated blood pressure Systolic ≥130 and/or diastolic ≥85 mm Hg;
Elevated fasting glucose ≥100 mg;


Note Some US adults of non-Asian origin (eg, white, black, Hispanic) with marginally increased waist circumference (eg,[37–39 inches] in men and [31–34 inches] in women) may have strong genetic contribution to insulin resistance and should benefit from changes in lifestyle habits, similar to men with categorical increases in waist circumference. Lower waist circumference cut point (eg,[35 inches] in men and (31 inches) in women) appears to be appropriate for Asian Americans.
Aso, certain drug treatments or Docter suppervised programs may alter the above guidelines.

Glycated haemoglobin:
2023 ESC Guidelines

Following high-quality epidemiological studies, it was suggested that HbA1c could be used to diagnose diabetes, and this was subsequently endorsed by international guidelines. It should be noted that epidemiological studies have relied on the adult population, though HbA1c is also used in younger individuals as a diagnostic test. Advantages of HbA1c include ease of measurement, limited within-individual variability, and the convenience of anytime testing without the need for fasting or a cumbersome OGTT.
However, HbA1c is not accurate in specific groups where the relationship between HbA1c and glucose levels is altered for any reason Moreover, in cases of shorter diabetes duration, such as early type 1 diabetes mellitus (T1DM) or acute pancreatic damage, HbA1c can lead to false-negative results. Another practical limitation is the lack of test availability in some parts of the world due to financial constraints.
Guidelines agree that HbA1c ≥48 mmol/mol (≥6.5%) is diagnostic of diabetes, while the diagnosis of pre-diabetes uses two different cut-off values. The WHO criteria define pre-diabetes as HbA1c 42–47 mmol/mol (6.0–6.4%), while the ADA recommends a wider range of 39–47 mmol/mol (5.7–6.4%). Notably, the combination of HbA1c and fasting glucose in the diabetes range is diagnostic of diabetes and a second test is not required, even if the individual is asymptomatic. However, if the two are discordant, the number in the diabetes range should be repeated or, preferably, an OGTT performed, which remains the gold standard for diagnosing diabetes in unclear cases. The criteria used for diagnosing diabetes and pre-diabetes are summarized in Table 6. It should be noted that data from 73 studies on 294 998 individuals without known diabetes suggest that HbA1c is as good as or better than fasting, random, or post-load glucose levels for predicting CV risk. Refer to the ESC 2023 guidelines below.



CELL17



Diagnostic Criteria for Metabolic Syndrome

VIDEO: The American Diabetes Association’s Standards of Care in Diabetes—2024

Diagnosis and Management of the Metabolic Syndrome

2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes: Developed by the task force on the management of cardiovascular disease in patients with diabetes of the European Society of Cardiology


If you Would you like to run a Metabolic Health Anaysis based on the Metabolic Syndrome with some information from your current lipid panel use this:

LINK to TESTS


BACK TO MENU


INSULIN, GLUCAGON & HYPERINSULINEMIA



INSULIN29

INSULIN8

Insulin is a peptide hormone secreted by the β cells of the pancreatic islets of Langerhans and maintains normal blood glucose levels by facilitating cellular glucose uptake, regulating carbohydrate, lipid and protein metabolism and promoting cell division and growth through its mitogenic effects.

Insulin resistance is defined where a normal or elevated insulin level produces an attenuated biological response; classically this refers to impaired sensitivity to insulin mediated glucose disposal.


INSULIN14

Compensatory hyperinsulinaemia occurs when pancreatic β cell secretion increases to maintain normal blood glucose levels in the setting of peripheral insulin resistance in muscle and adipose tissue.

In most natural habitats, calorie availability is scarce and unpredictable, necessitating the evolution of systems for the efficient storage and utilization of energy. But in our modern, mechanized society, caloric demands are minimized, while highly palatable, calorie-dense foods and beverages are readily available. These changes have fostered the current pandemic of obesity and comorbid conditions of nonalcoholic fatty liver disease (NAFLD), atherosclerosis, and type 2 diabetes (T2D).

Here is a possible representation of the relationship between hyperinsulinemia and fasting hyperglycemia:
Years 1-5: Insulin resistance increases, leading to hyperinsulinemia (≥25 mIU/ml) with normal Glucose levels.
Years 5-10: Hyperinsulinemia persists, and insulin resistance worsens, leading to impaired glucose tolerance and prediabetes: ( 100-125mg/dL or 5.6-6.9 mmol/L)
Years 10-15: Fasting hyperglycemia (≥126 mg/dL or 6.9 mmol/L) develops, indicating the onset of Type 2 diabetes


T2D1B



Insulin maintains normal glucose levels despite increasing insulin resistance through a process called compensatory hyperinsulinemia.
Increased Insulin Secretion: As insulin resistance develops, the body’s cells (particularly muscle, fat, and liver cells) become less responsive to insulin. To compensate for this reduced sensitivity, the pancreas produces and secretes more insulin. This increased insulin production helps to maintain normal blood glucose levels by ensuring that glucose can still be taken up by cells for energy or storage
Enhanced Beta Cell Function: The pancreatic beta cells, which are responsible for producing insulin, work harder and become more efficient at secreting insulin in response to rising blood glucose levels. This enhanced function helps to counteract the effects of insulin resistance
Suppression of Hepatic Glucose Production: Insulin plays a crucial role in suppressing the liver’s production of glucose. With higher levels of insulin, the liver’s glucose output is reduced, which helps to keep blood glucose levels within the normal range.
Increased Insulin Sensitivity in Other Tissues: While insulin resistance primarily affects muscle, fat, and liver cells, other tissues in the body may remain more sensitive to insulin. This differential sensitivity helps to maintain overall glucose homeostasis.
Feedback Mechanisms: The body has several feedback mechanisms that help regulate insulin and glucose levels. For example, when blood glucose levels rise, the pancreas releases more insulin. Conversely, when blood glucose levels fall, insulin secretion decreases. These feedback loops help to maintain glucose levels within a narrow range.
Despite these compensatory mechanisms, over time, the pancreas may become unable to produce enough insulin to overcome the increasing resistance. When this happens, blood glucose levels start to rise, leading to conditions such as Impaired Fasting Glucose (IFG) and eventually type 2 diabetes.


T2D2






Role of PI3K/AKT Pathway in Insulin-Mediated Glucose Uptake

The pathogenesis of insulin resistance: integrating signaling pathways and substrate flux

Prediabetes: A high-risk state for developing diabetes

Hyperinsulinemia: an early biomarker of metabolic dysfunction

Trajectories of Glycemia, Insulin Sensitivity and Insulin Secretion Preceding the Diagnosis of Type 2 Diabetes: The Whitehall II Study

Insulin Resistance and Hyperinsulinemia

Hyperinsulinemia in Obesity, Inflammation, and Cancer

Insulin Preferentially Regulates the Activity of Parasympathetic Preganglionic Neurons over Sympathetic Preganglionic Neurons

Hyperglycaemia reduces gastrin-stimulated gastric acid secretion in humans

Hyperinsulinemia and Its Pivotal Role in Aging, Obesity, Type 2 Diabetes, Cardiovascular Disease and Cancer

β Cell GHS-R Regulates Insulin Secretion and Sensitivity

Hepatic Insulin Clearance: Mechanism and Physiology

Hyperglycemia alters PI3k and Akt signaling and leads to endothelial cell proliferative dysfunction

Hyperinsulinemia: An Early Indicator of Metabolic Dysfunction

Molecular Physiology of Insulin Function

Physiology of insulin

Overview of Angiogenesis

Insulin and Insulin Resistance

Mechanisms of Insulin Action and Insulin Resistance

Hyperinsulinemia and Its Pivotal Role in Aging, Obesity, Type 2 Diabetes, Cardiovascular Disease and Cancer

A Receptor Story: Insulin Resistance Pathophysiology and Physiologic Insulin Resensitization’s Role as a Treatment Modality

The Insulin Receptor and Its Signal Transduction Network

Activation mechanism of the insulin receptor: a structural perspective

Does the association of the triglyceride to high-density lipoprotein cholesterol ratio with fasting serum insulin differ by race/ethnicity?

Triglyceride/HDL-Cholesterol Ratio as an Index of Intracranial Atherosclerosis in Nonstroke Individuals

The Triglyceride/High-Density Lipoprotein Cholesterol (TG/HDL-C) Ratio as a Risk Marker for Metabolic Syndrome and Cardiovascular Disease

The visceral adiposity index is a predictor of incident nonalcoholic fatty liver disease:

Triglyceride glucose index for predicting cardiovascular outcomes in patients with coronary artery disease

Changes in Cells Associated with Insulin Resistance

Targeting hepatic glucose output in the treatment of type 2 diabetes

Pathogenesis of Insulin Resistance in Skeletal Muscle

The pathogenesis of insulin resistance: integrating signaling pathways and substrate flux

Selective Insulin Resistance and the Development of Cardiovascular Diseases in Diabetes: The 2015 Edwin Bierman Award Lecture

Hyperglycemia- and hyperinsulinemia-induced insulin resistance causes alterations in cellular bioenergetics and activation of inflammatory signaling in lymphatic muscle

Insulin Signaling And Insulin Resistance

Lipid-induced insulin resistance: unravelling the mechanism

Ectopic Fat Accumulation in Distinct Insulin Resistant Phenotypes; Targets for Personalized Nutritional Interventions

Ectopic Fat and Insulin Resistance: Pathophysiology and Effect of Diet and Lifestyle Interventions

Insulin Degradation: Progress and Potential*

Role of Insulin Clearance in Insulin Action and Metabolic Diseases

The Physiology of Insulin Clearance

Insulin Resistance and Hyperinsulinemia: Is hyperinsulinemia the cart or the horse?

Effect of Intermittent Fasting Diet on Glucose and Lipid Metabolism and Insulin Resistance in Patients with Impaired Glucose and Lipid Metabolism

Cephalic phase insulin release: A review of its mechanistic basis and variability in humans

Glucose‐stimulated insulin secretion: A newer perspective

The Hidden Problem of Chronic Hyperinsulinemia

Mechanisms of muscle insulin resistance and the cross-talk with liver and adipose tissue

Hyperinsulinemia alters insulin receptor presentation and internalization in brain microvascular endothelial cells

The Roles of the IGF Axis in the Regulation of the Metabolism: Interaction and Difference between Insulin Receptor Signaling and IGF-I Receptor Signaling

The growth hormone–insulin-like growth factor-I axis in the diagnosis and treatment of growth disorders

Insulin-like Growth Factor and its Therapeutic Potential for Diabetes Complications - Mechanisms and Metabolic Links:

Recent Advances in Our Understanding of Insulin Action and Insulin Resistance

The Multifunctionality of CD36 in Diabetes Mellitus and Its Complications—Update in Pathogenesis, Treatment and Monitoring

Insulin–Heart Axis: Bridging Physiology to Insulin Resistance

Insulin signalling and the regulation of glucose and lipid metabolism

Insulin action and resistance in obesity and type 2 diabetes

On the causal relationships between hyperinsulinaemia, insulin resistance, obesity and dysglycaemia in type 2 diabetes

Early beta cell dysfunction vs insulin hypersecretion as the primary event in the pathogenesis of dysglycaemia

Diabetes: Have We Got It All Wrong?

Hyperinsulinemia and Its Pivotal Role in Aging, Obesity, Type 2 Diabetes, Cardiovascular Disease and Cancer

Prediabetes: A high-risk state for developing diabetes

Insulin Resistance Mayo Clinic

Hyperinsulinemia: An Early Indicator of Metabolic Dysfunction

Current Studies on Molecular Mechanisms of Insulin Resistance

Insulin: too much of a good thing is bad

The pathogenesis of insulin resistance: integrating signaling pathways and substrate flux

Insulin Resistance

Mechanisms of Insulin Action and Insulin Resistance

Defining and Characterizing the Progression of Type 2 Diabetes

Mechanisms of β-Cell Death in Type 2 Diabetes

The role of interleukin-1β in type 2 diabetes mellitus: A systematic review and meta-analysis

Insulin and aging – a disappointing relationship

Probing the Relationship Between Insulin Sensitivity and Longevity Using Genetically Modified Mice

Insulin Resistance: The Increased Risk of Cancers

Plant-Based Diet Indices and Their Association with Frailty in Older Adults

Assessment HOMA as a predictor for new onset diabetes mellitus and diabetic complications in non-diabetic adults: a KoGES prospective cohort study

Diet-Induced Hyperinsulinemia as a Key Factor in the Etiology of Both Benign Prostatic Hyperplasia and Essential Hypertension?

Linking Pre-Diabetes with Benign Prostate Hyperplasia. IGFBP-3: A Conductor of Benign Prostate Hyperplasia Development Orchestra?

Endocrinology of the Aging Prostate: Current Concepts

Alterations of Gut Microbiota by Overnutrition Impact Gluconeogenic Gene Expression and Insulin Signaling

https://pmc.ncbi.nlm.nih.gov/articles/PMC9135930/

Dopamine Negatively Regulates Insulin Secretion

Insulin resistance in brain alters dopamine turnover and causes behavioral disorders

Insulin Regulates Brain Function, but How Does It Get There

The Role of Hypothalamic Microglia in the Onset of Insulin Resistance and Type 2 Diabetes: A Neuro-Immune Perspective

Does Insulin Play a Role in Prostate Cancer

Fasting Insulin and Risk of Cancer Related Mortality in Non-diabetic Adults

A review of the carbohydrate–insulin model of obesity

The Carbohydrate-Insulin Model of Obesity: Beyond "Calories In, Calories Out"

Relationship Between Insulin Resistance and an Endogenous Nitric Oxide Synthase Inhibitor

Hyperinsulinemia in Obesity, Inflammation, and Cancer

Diabetes: Have We Got It All Wrong?

Regulation of insulin secretion: a matter of phase control and amplitude modulation

Paracrine regulation of insulin secretion

Abnormal pancreatic glucagon secretion and postprandial hyperglycemia in diabetes mellitus

The Difference δ-Cells Make in Glucose Control


INSULIN RECEPTOR

Insulin Receptor Trafficking: Consequences for Insulin Sensitivity and Diabetes

Insulin Receptor Signaling in Normal and Insulin-Resistant States

The role of GLUT2 in glucose metabolism in multiple organs and tissues

The facilitative glucose transporter GLUT3: 20 years of distinction

Glucose transporters in pancreatic islets

Hypertonicity during a rapid rise in D-glucose mediates first-phase insulin secretion

Glucose transporters in pancreatic islets

Current understanding of glucose transporter 4 expression and functional mechanisms

Regulation of insulin receptor function

Regulation of insulin sensitivity by serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2

The Insulin Receptor and Its Signal Transduction Network

Insulin Receptor Signaling in Normal and Insulin-Resistant States

Novel roles for insulin receptor (IR) in adipocytes and skeletal muscle cells via new and unexpected substrates

The dynamic clustering of insulin receptor underlies its signaling and is disrupted in insulin resistance

Molecular Mechanisms of Insulin Resistance: Serine Phosphorylation of Insulin Receptor Substrate-1 and Increased Expression of p85α: The Two Sides of a Coin

Agonism and Antagonism at the Insulin Receptor


Ast Alt Ratio Calculator


BACK TO MENU


GLUCOSE & HYPERGLYCEMIA

The term "Hyperglycemia" is derived from the Greek hyper (high) + glykys (sweet/sugar) + haima (blood). Hyperglycemia is defined as blood glucose greater than 125 mg/dL while fasting and greater than 180 mg/dL 2 hours postprandial. A patient has impaired glucose tolerance, or pre-diabetes, with a fasting plasma glucose of 100 mg/dL to 125 mg/dL.

Chronic exposure to a glucose rich environment creates several physiological and pathophysiological changes. There are several pathways by which hyperglycemia exacerbate its toxic effect on cells, tissues and organ systems. Hyperglycemia can induce oxidative stress, upsurge polyol pathway, activate protein kinase C (PKC), enhance hexosamine biosynthetic pathway (HBP), promote the formation of advanced glycation end-products (AGEs) and finally alters gene expressions. Prolonged hyperglycemic condition leads to severe diabetic condition by damaging the pancreatic β cell and inducing insulin resistance. I hope to cover most of these concerns with the appropriate studies and papers, some published and some in process. The idea is to try and understand what hyperglycemia and hyperinsuinemia do to the different pathways and body organs. I have found that this is a task beyond my comprehension, but there are studies that I can relate to and can help others to appreciate.



GLU13



- Pathogenesis of Chronic Hyperglycemia: From Reductive Stress to Oxidative Stress

Prediabetes: A high-risk state for developing diabetes

Trajectories of Glycemia, Insulin Sensitivity and Insulin Secretion Preceding the Diagnosis of Type 2 Diabetes:

The new biology of diabetes

VIDEO

MAYO CLINIC: Hyperglycemia,

ADA: Hyperglycemia (High Blood Glucose)

Association of Glycemic Indices (Hyperglycemia, Glucose Variability, and Hypoglycemia) with Oxidative Stress and Diabetic Complications

Vascular Dysfunction in Hyperglycemia

Chronic hyperglycemia mediated physiological alteration and metabolic distortion leads to organ dysfunction, infection, cancer progression and other pathophysiological consequences: An update on glucose toxicity

ABSTRACT: Hyperglycemia, lipoprotein glycation, and vascular disease

VIDEO: The Effects of Hyperglycemia on the Immune System

Hyperglycemia

Historical background

Diabetes and mitochondrial function: Role of hyperglycemia and oxidative stress

Circadian Regulation of Glucose, Lipid, and Energy Metabolism in Humans

Regulation of glucose metabolism from a liver-centric perspective

Fasting plasma glucose levels and coronary artery calcification in subjects with impaired fasting glucose

Impact of HbA1c Testing at Point of Care on Diabetes Management

A Practical Review of C-Peptide Testing in Diabetes

Hyperglycemia and Oxidative Stress: An Integral, Updated and Critical Overview of Their Metabolic Interconnections

Glucotoxicity promotes aberrant activation and mislocalization of Ras-related C3 botulinum toxin substrate 1 [Rac1] and metabolic dysfunction in pancreatic islet β-cells: Reversal of such metabolic defects by metformin

The Role of CD36 in Type 2 Diabetes Mellitus: β-Cell Dysfunction and Beyond

Effect of hyperglycemia on gastric acid secretion during the gastric phase of digestion

The Relationship between Erythrocytes and Diabetes Mellitus

Effect of high glucose concentrations on human erythrocytes in vitro

Type 2 Diabetes Mellitus: A Pathophysiologic Perspective

Updates in the Management of Hyperglycemic Crisis

Relationships BetweenGastric Emptying, Postprandial Glycemia,and Incretin Hormone

Postprandial Hyperlipidemia: Its Pathophysiology, Diagnosis, Atherogenesis, and Treatments

Effect of acute hypohydration on glycemic regulation in healthy adults

The Effect of Short-Term Hyperglycemia on the Innate Immune System

Characteristics of glucose transporters

Hyperglycemia- and hyperinsulinemia-induced insulin resistance causes alterations in cellular bioenergetics and activation of inflammatory signaling in lymphatic muscle

Role of insulin and other related hormones in energy metabolism

INSULIN, MUSLE UPTAKE, AND HEXOKINASE

VIDEO: The Effects of Hyperglycemia on the Immune System

VIDEO: How diabetes destroys the human body

VIDEO: Hyperglycemia and neuropathy, nephropathy and retinopathy. DM complications

Mechanisms underlying the pathophysiology of type 2 diabetes: From risk factors to oxidative stress, metabolic dysfunction, and hyperglycemia

Potential Role of Protein Kinase C in the Pathophysiology of Diabetes-Associated Atherosclerosis

Impaired glucose tolerance and impaired fasting glucose share similar underlying pathophysiologies

The Promising Frontier of Cardiometabolic Syndrome: A New Paradigm in Cardiology

Gluconeogenesis

p38 Mitogen-activated Protein Kinase Plays a Stimulatory Role in Hepatic Gluconeogenesis

The Aging Vasculature: Glucose Tolerance, Hypoglycemia and the Role of the Serum Response Factor

Age-related Changes in Glucose Metabolism, Hyperglycemia, and Cardiovascular Risk

Metabolic flux and the regulation of mammalian cell growth

Pancreatic regulation of glucose homeostasis

Insulin, Muscle Glucose Uptake, and Hexokinase:

Regulation of GLUT4 and Insulin-Dependent Glucose Flux

Repeated glucose spikes and insulin resistance synergistically deteriorate endothelial function

Hexokinase-2-Linked Glycolytic Overload and Unscheduled Glycolysis—Driver of Insulin Resistance and Development of Vascular Complications of Diabetes

Some Details of Glycolysis


GLYCOLYSIS

GLY8

Glycolysis represents the process of breaking down monosaccharides, which involves the energy metabolism, homeostasis, and the linkage of various physiological functions such as muscle movement, development, neurotransmission, etc. what happens when there are Elevated levels of ATP and alanine create negative feedback to inhibit PK, does the flow reverese bak to g6p When there are elevated levels of ATP and alanine, they act as negative feedback inhibitors for pyruvate kinase (PK). This inhibition slows down the conversion of phosphoenolpyruvate (PEP) to pyruvate in glycolysis. As a result, the flow of metabolites is redirected upstream.

However, the flow does not reverse back to glucose-6-phosphate (G6P). Instead, the accumulation of PEP and other intermediates can lead to an increase in the levels of fructose-1,6-bisphosphate (F1,6BP), which can then be converted back to fructose-6-phosphate (F6P) and eventually to G6P through gluconeogenesis or other pathways

Phosphofructokinase is the main enzyme controlled in glycolysis. High levels of ATP, citrate, or a lower, more acidic pH decrease the enzyme’s activity. An increase in citrate concentration can occur because of a blockage in the citric acid cycle. Fermentation, with its production of organic acids like lactic acid, frequently accounts for the increased acidity in a cell; however, the products of fermentation do not typically accumulate in cells.


GLYCONEOGENESIS


Glycolysis and Gluconeogenesis - Reciprocal Regulation

The neglected PCK1/glucagon (inter)action in nutrient homeostasis beyond gluconeogenesis

Diabetic-induced alterations in hepatic glucose and lipid metabolism:


GLYCOGENOLESIS

Glucagon is a hormone produced by the alpha cells of the pancreas and plays a key role in regulating blood sugar levels. Glucagon acts in opposition to insulin: while insulin promotes glucose uptake and storage, glucagon promotes the breakdown of glycogen stores in the liver and the release of glucose into the bloodstream.

The switch from insulin secretion to glucagon secretion typically occurs when blood glucose levels start to decrease. The transition between insulin and glucagon secretion is tightly regulated to maintain blood sugar balance. In general, glucagon secretion is stimulated when blood glucose levels drop below a certain threshold. The approximate threshold for glucagon secretion is around 70-80 mg/dL (3.9-4.4 mmol/L) of blood glucose. When blood glucose levels decrease below this threshold, the pancreas responds by reducing insulin secretion and increasing glucagon secretion to promote the release of glucose from glycogen stores in the liver.

During periods of fasting, prolonged exercise, or low-carbohydrate intake, blood glucose levels can decrease, triggering glucagon secretion to maintain blood sugar levels and provide energy for the body's cells. Overall, the switch from insulin secretion to glucagon secretion occurs in response to decreasing blood glucose levels and is an essential mechanism for maintaining blood sugar balance and supporting energy needs during fasting or periods of increased demand.

Glucokinase intrinsically regulates glucose sensing and glucagon secretion in pancreatic alpha cells

Glucagon Physiology

Glucagon

Glucose Controls Glucagon Secretion by Regulating Fatty Acid Oxidation in Pancreatic α-Cells

The Regulation and Secretion of Glucagon in Response to Nutrient Composition

Role of GLUT1 in regulation of reactive oxygen species

Physiology, Glucagon

Glucagon Signaling Pathway

Glucagon Receptor Signaling and Glucagon Resistance

Insulin Resistance Is Accompanied by Increased Fasting Glucagon and Delayed Glucagon Suppression in Individuals With Normal and Impaired Glucose Regulation

Markers of Glucagon Resistance Improve With Reductions in Hepatic Steatosis and Body Weight in Type 2 Diabetes

The Vicious Circle of Hepatic Glucagon Resistance in Non-Alcoholic Fatty Liver Disease

Glucagon Receptor Signaling and Glucagon Resistance

Insulin Resistance Is Accompanied by Increased Fasting Glucagon and Delayed Glucagon Suppression in Individuals With Normal and Impaired Glucose Regulation


GLYCONEOGENOLYSIS



GLUT TRANSPORTERS


Glucose transporters: physiological and pathological roles

Glucose transporter 1 in health and disease

Functional Properties and Genomics of Glucose Transporters

INSULIN, MUSLE UPTAKE, AND HEXOKINASE

Skeletal Muscle GLUT1 Transporter Protein Expression and Basal Leg Glucose Uptake Are Reduced in Type 2 Diabetes

Glucose transport and sensing in the maintenance of glucose homeostasis and metabolic harmony

Glucose transporters in pancreatic islets

The role of GLUT2 in glucose metabolism in multiple organs and tissues

The facilitative glucose transporter GLUT3: 20 years of distinction

Glucose transporters in pancreatic islets

Glucose transporters in pancreatic islets

Current understanding of glucose transporter 4 expression and functional mechanisms

Importance of GLUT Transporters in Disease Diagnosis and Treatment

GLUT5: structure, functions, diseases and potential applications

The role of GLUT2 in glucose metabolism in multiple organs and tissues

ABSTRACT ONLY: Regulation of glucose transport by insulin: traffic control of GLUT4

GLUT2, glucose sensing and glucose homeostasis

Glucose transporters in adipose tissue, liver, and skeletal muscle in metabolic health and disease

Pretranslational Suppression of a Glucose Transporter Protein Causes Insulin Resistance in Adipocytes from Patients with Non-Insulin-dependent Diabetes Mellitus and Obesity

Metabolic changes in aging humans

Alternative routes to the cell surface underpin insulin-regulated membrane trafficking of GLUT4

Insulin signalling and GLUT4 trafficking in insulin resistance

Glucose transporters in adipose tissue, liver, and skeletal muscle in metabolic health and disease

Palmitate-induced insulin resistance causes actin filament stiffness and GLUT4 mis-sorting without altered Akt signalling


BACK TO MENU


Diet & DIGESTION

OPEN

Effect of hyperglycemia on gastric acid secretion during the gastric phase of digestion

Alcohol and gastric acid secretion in humans.

Effect of alterations of blood glucose levels on gastric acid secretion, plasma gastrin, and plasma osmolality in man

Neuroendocrine control of appetite and metabolism

Autonomic control of energy balance and glucose homeostasis

Fat Digestion - Lipolysis & Lipid Transport DM

Omega-6 vegetable oils as a driver of coronary heart disease: the oxidized linoleic acid hypothesis

Activation of G protein-coupled receptors by ketone bodies: Clinical implication of the ketogenic diet in metabolic disorders

Effect of weight loss on the rate of muscle protein synthesis during fasted and fed conditions in obese older adults

Mediterranean Diet and Musculoskeletal-Functional Outcomes in Community-Dwelling Older People: A Systematic Review and Meta-Analysis

Effect of Protein Supplementation Combined With Resistance Training on Muscle Mass, Strength and Function in the Elderly

Insulin Resistance of Protein Metabolism in Type 2 Diabetes and Impact on Dietary Needs:

Effects of Dietary Starch Structure on Growth Performance, Serum Glucose–Insulin Response, and Intestinal Health in Weaned Piglets

Vitamins Important for Metabolism


BACK TO MENU




OVERNUTRITION

OVERNUTRITITION

A high-fat, Western-style diet is an important predisposing factor for the onset of type 2 diabetes and obesity. It causes changes in gut microbial profile, reduction of microbial diversity, and the impairment of the intestinal barrier, leading to increased serum lipopolysaccharide (endotoxin) levels. Elevated lipopolysaccharide (LPS) induces acetyltransferase P300 both in the nucleus and cytoplasm of liver hepatocytes through the activation of the IRE1-XBP1 pathway in the endoplasmic reticulum stress. In the nucleus, induced P300 acetylates CRTC2 to increase CRTC2 abundance and drives Foxo1 gene expression, resulting in increased expression of the rate-limiting gluconeogenic gene G6pc and Pck1 and abnormal liver glucose production. Furthermore, abnormal cytoplasm-appearing P300 acetylates IRS1 and IRS2 to disrupt insulin signaling, leading to the prevention of nuclear exclusion and degradation of FOXO1 proteins to further exacerbate the expression of G6pc and Pck1 genes and liver glucose production. Inhibition of P300 acetyltransferase activity by chemical inhibitors improved insulin signaling and alleviated hyperglycemia in obese mice. Thus, P300 acetyltransferase activity appears to be a therapeutic target for the treatment of type 2 diabetes and obesity. Refer to Source Here

OVERNUTRITITION
IRS acetylation by abnormal cytoplasm-appearing P300 causes insulin resistance.
(A) Insulin-mediated activation of PI3K-AKT signaling leads to FOXO1 phosphorylation, nuclear exclusion and degradation, subsequently inhibition of gluconeogenic gene expression in the liver.
(B) Overnutrition induced abnormal cytoplasm-appearing P300 acetylates IRS1 and IRS2 to disrupt their association with insulin receptors and insulin signaling. FOXO1 cannot be phosphorylated by AKT
(C), leading to its nuclear accumulation and stimulation of overexpression of the gluconeogenic gene in the liver
(D). The solid arrows indicate the direct effects, the dashed arrows indicate indirect effects, and the crossed line indicates the blockade of the pathway.
Refer to Source Here


_
Overnutrition, particularly a diet high in fructose (from sources like high-fructose corn syrup and sucrose), high glycemic load carbohydrates, and fat should cause a metabolic cascade of:
1. Lipogenesis:
2. De Novo Lipolysis:
3. De Novo Gluconeogenesis as Overnutrition should cause regulation if glycolysis:
4. Elevated levels of Palmitate:
5. Hyperglycemia:
6. Hyperinsulinemia:
7. Increased levels of P300 in the liver through the leaky gut.
8. Insulin resistance in the liver directly:
9. Huge Metabolic problems:
10. This should cause cytokines to be circulated, IL6 etc


Alterations of Gut Microbiota by Overnutrition Impact Gluconeogenic Gene Expression and Insulin Signaling

p300 or CBP is required for insulin-stimulated glucose uptake in skeletal muscle and adipocytes

The carbohydrate-insulin model: a physiological perspective on the obesity pandemic

The Impact of Overnutrition on Insulin Metabolic Signaling in the Heart and the Kidney

Assessing the Nutrient Composition of a Carnivore Diet:

Basic concepts in nutrition: Overnutrition – Functional and clinical consequences

Overnutrition causes insulin resistance and metabolic disorder through increased sympathetic nervous system activity

Neuroinflammation and Neurodegeneration in Overnutrition-induced Diseases


BACK TO MENU




METABOLISM

METABOLISM SLIDESHOW

Metabolism

Enzymes: principles and biotechnological applications

The physiological regulation of glucose flux into muscle in vivo

Insulin and β adrenergic receptor signaling: Cross talk in heart

The Multifaceted Pyruvate Metabolism: Role of the Mitochondrial Pyruvate Carrier

Regulation of Glycolysis

AMINO ACID METABOLISM1

AMINO ACID METABOLISM2

Metabolic flexibility and insulin resistance

Metabolism

Glucose transporters: physiological and pathological roles

Evidence for Defects in the Trafficking and Translocation of GLUT4 Glucose Transporters in Skeletal Muscle as a Cause of Human Insulin Resistance

Protein Metabolism

Michaelis Constant (Km)

10 STEPS OF GLYCOLISIS

8 STEPS OF THE TCA CYCLE

Amino Acid Metabolism

VIDEO: Steps of Glycolysis Reactions Explained - Animation - SUPER EASY

Importance of GLUT Transporters in Disease Diagnosis and Treatment

The Metabolic Map: Proteins

Metabolism | The Metabolic Map: Carbohydrates

Metabolism | The Metabolic Map: Lipids

Metabolism | Glycolysis

Metabolism | Regulation of Glycolysis

Brain and systemic glucose metabolism in the healthy elderly following fish oil supplementation

Molecular Pathophysiology of Hepatic Glucose Production

The Liver & Blood Sugar

REGULATION OF GLUCOSE PRODUCTION BY THE LIVER

Relationship between Added Sugars Consumption and Chronic Disease Risk Factors: Current Understanding

Intakes and sources of dietary sugars and their association with metabolic and inflammatory markers

Sucrose

Protein Metabolism

Lipid Metabolism

Vitamins and Minerals Involved in Energy Metabolism

Glutathione Stimulates Vitamin D Regulatory and Glucose-Metabolism Genes, Lowers Oxidative Stress and Inflammation, and Increases 25-Hydroxy-Vitamin D Levels in Blood:

Beta-Aminoisobutyric Acid as a Novel Regulator of Carbohydrate and Lipid Metabolism

The Role of Glutathione in Protecting against the Severe Inflammatory Response Triggered by COVID-19

Glutathione Stimulates Vitamin D Regulatory and Glucose-Metabolism Genes, Lowers Oxidative Stress and Inflammation, and Increases 25-Hydroxy-Vitamin D Levels in Blood:

Metabolic Stability of D-Allulose in Biorelevant Media and Hepatocytes: Comparison with Fructose and Erythritol

Serum glucose and insulin levels and erythritol balance after oral administration of erythritol in healthy subjects

Erythritol Attenuates Postprandial Blood Glucose by Inhibiting α-Glucosidase

D-ALLOUSE Comparison with Fructose and Erythritol

AMP kinase regulation of sugar transport in brain capillary endothelial cells during acute metabolic stress

High-Fat Diet or Diabetes Drug May Enhance Response to Targeted Cancer Drug

Carbotoxicity—Noxious Effects of Carbohydrates

Differentiation of Diabetes by Pathophysiology, Natural History, and Prognosis

A Fresh View of Glycolysis and Glucokinase Regulation: History and Current Status*

Fatty acid metabolism, energy expenditure and insulin resistance in muscle

The role of fatty acids in insulin resistance

Evidence for Central Regulation of Glucose Metabolism

Peroxisome Proliferator-Activated Receptors and Caloric Restriction—Common Pathways Affecting Metabolism, Health, and Longevity

AMP-activated protein kinase: Ancient energy gauge provides clues to modern understanding of metabolism

Endothelial GLUTs and vascular biology

Neurotransmitters in Type 2 Diabetes and the Control of Systemic and Central Energy Balance

Brain control of blood glucose levels: implications for the pathogenesis of type 2 diabetes

Brain control of blood glucose levels: implications for the pathogenesis of type 2 diabetes

Autonomic control of energy balance and glucose homeostasis

Cardiometabolic multimorbidity, lifestyle behaviours, and cognitive function

Autonomic Regulation of Glucose Homeostasis: a Specific Role for Sympathetic Nervous System Activation

Sympathetic nervous system and immune interplay: Key to metabolic regulation

Relationship Between Glucocorticoids and Insulin Resistance in Healthy Individuals

A Mechanistic Review on How Berberine Use Combats Diabetes and Related Complications: Molecular, Cellular, and Metabolic Effects

Dietary Strategies for Improving Post-Prandial Glucose, Lipids, Inflammation, and Cardiovascular Health

Diabetes and Renin-Angiotensin-Aldosterone System: Pathophysiology and Genetics

Metabolism | Glycolysis NN

Metabolism and Urea Cycle

Metabolism Mobilization of Triglycerides NN

Metabolism Overview DR MIKE

Protein Metabolism

Glucose Ketone Index (GKI) Calculator

Lipid Metabolism

The Randle cycle revisited: a new head for an old hat

The Biochemistry and Physiology of Mitochondrial Fatty Acid β-Oxidation and Its Genetic Disorders

Amino Acid Metabolism

Lipid and glucose metabolism in senescence

Circadian Syndrome Is Associated with Dietary Patterns among Middle-Older Americans

Chemical Reactions in Living Things

Muscle Protein Synthesis and Whole-Body Protein Turnover Responses to Ingesting Essential Amino Acids, Intact Protein, and Protein-Containing Mixed Meals with Considerations for Energy Deficit

Regulation of Glycolysis

Metabolic landscape in cardiac aging: insights into molecular biology and therapeutic implications

Metabolic changes in aging humans: current evidence and therapeutic strategies

METABOLIC FLEXABILITY

OPEN

The Effects of Ketogenic Diet on Insulin Sensitivity and Weight Loss,

Targeting the fatty acid transport proteins (FATP) to understand the mechanisms linking fatty acid transport to metabolism

Metabolic Flexibility as an Adaptation to Energy Resources and Requirements in Health and Disease

Metabolic Flexibility and Its Impact on Health Outcomes

VIDEO Protein Metabolism Overview, Animation

BACK TO MENU


PI3K-AKT PATHWAY

PAN11



The PI3K/AKT pathway in obesity and type 2 diabetes

Role of PI3K/AKT Pathway in Insulin-Mediated Glucose Uptake

Insulin–PI3K signalling: an evolutionarily insulated metabolic driver of cancer

Akt2 ablation prolongs life span and improves myocardial contractile function with adaptive cardiac remodeling: role of Sirt1‐mediated autophagy regulation

Autophagy as an Emerging Target in Cardiorenal Metabolic Disease: from Pathophysiology to Management

VIDEO PTEN regulation

Metabolic Role of PTEN in Insulin Signaling and Resistance

Inhibition of AMPK and Krebs cycle gene expression drives metabolic remodeling of Pten-deficient preneoplastic thyroid cells

Metabolism and proliferation share common regulatory pathways in cancer cells

The PI3K/AKT pathway in obesity and type 2 diabetes.

Obesity enhances nongenomic estrogen receptor crosstalk with the PI3K/Akt and MAPK pathways to promote in vitro measures of breast cancer progression

TCF7L2 regulates pancreatic β-cell function through PI3K/AKT signal pathway

The PTEN–PI3K pathway: of feedbacks and cross-talks

The Role of PKC-MAPK Signalling Pathways in the Development of Hyperglycemia-Induced Cardiovascular Complications

MAPK signal pathways in the regulation of cell proliferation in mammalian cells

Loss of mTORC1 signaling alters pancreatic α cell mass and impairs glucagon secretion

Berberine Slows the Progression of Prediabetes to Diabetes in Zucker Diabetic Fatty Rats by Enhancing Intestinal Secretion of Glucagon-Like Peptide-2 and Improving the Gut Microbiota

IRS1/PI3K/AKT pathway signal involved in the regulation of glycolipid metabolic abnormalities

Altered Insulin Signaling in Alzheimer’s Disease Brain – Special Emphasis on PI3K-Akt Pathway

The PI3K pathway in human disease

Transcriptional Regulation of INSR, the Insulin Receptor Gene

Signaling pathways in insulin action: molecular targets of insulin resistance

Role of PI3K/AKT Pathway in Insulin-Mediated Glucose Uptake

The role of skeletal muscle Akt in the regulation of muscle mass and glucose homeostasis

An Integrated View of Insulin Resistance and Endothelial Dysfunction

AKT/PKB Signaling: Navigating Downstream

PTEN and the PI3-Kinase Pathway in Cancer

PTEN function, the long and the short of it

Chronic and Intermittent Hyperglycemia Modulates Expression of Key Molecules of PI3K/AKT Pathway in Differentiating Human Visceral Adipocytes

Hyperglycemia alters PI3k and Akt signaling

Regulation of PTEN translation by PI3K signaling maintains pathway homeostasis

Management of Phosphatidylinositol-3-Kinase Inhibitor-Associated Hyperglycemia

VIDEO PI3K/Akt pathway - part 5: PTEN

Molecular Targeting of the Phosphoinositide-3-Protein Kinase (PI3K) Pathway across Various Cancers

Phosphatidylinositol-4-Phosphate-5-Kinase α Deficiency Alters Dynamics of Glucose-Stimulated Insulin Release to Improve Glucohomeostasis and Decrease Obesity in Mice

The PTEN/PI3K/AKT Pathway in vivo, Cancer Mouse Models

Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation

Loss of PTEN expression is associated with PI3K pathway-dependent metabolic reprogramming in hepatocellular carcinoma

The PTEN–PI3K pathway: of feedbacks and cross-talks

PTEN Inhibition in Human Disease Therapy

Metabolic Role of PTEN in Insulin Signaling and Resistance

The Impact of PIK3R1 Mutations and Insulin–PI3K Glycolytic Pathway Regulation in Prostate Cancer

find

PTEN Mutations as a Cause of Constitutive Insulin Sensitivity and Obesity

https://journals.physiology.org/doi/epdf/10.1152/ajpheart.01088.2004

How does hepatic lipid accumulation lead to lipotoxicity in non-alcoholic fatty liver disease

Regulation of Energy Metabolism by Receptor Tyrosine Kinase Ligands

The association of phosphoinositide 3-kinase enhancer A with hepatic insulin receptor enhances its kinase activity

Targeting PI3K/AKT signaling pathway in obesity

Identification of a leucine-mediated threshold effect governing macrophage mTOR signalling and cardiovascular risk

VIDEO SUSANNAH HANNAFORD Insulin PI3K Akt signaling pathway

Role of PI3K/AKT Pathway in Insulin-Mediated Glucose Uptake

3. Role of PI3K/AKT Pathway in Insulin-Mediated Glucose Uptake

The PI3K/AKT pathway in obesity and type 2 diabetes



The Critical Role of Akt in Cardiovascular Function

Molecular Targeting of the Phosphoinositide-3-Protein Kinase (PI3K) Pathway across Various Cancers

Insulin Inhibits Lipolysis in Adipocytes via the Evolutionarily Conserved mTORC1-Egr1-ATGL-Mediated Pathway

PI3K-AKT Signaling Pathway

Targeting PI3K/Akt signal transduction for cancer therapy

Leucine and mTORc1 act independently to regulate 2-deoxyglucose uptake in L6 myotubes

Amino acid-dependent control of mTORC1 signaling: a variety of regulatory modes

Obesity Alters the Muscle Protein Synthetic Response to Nutrition and Exercise

Muscle Protein Synthesis and Whole-Body Protein Turnover Responses to Ingesting Essential Amino Acids, Intact Protein, and Protein-Containing Mixed Meals with Considerations for Energy Deficit

VIDEO BITTMAN MTOR C1

VIDEO BITTMAN Sarcopenic Obesity

AMPK: Mechanisms of Cellular Energy Sensing and Restoration of Metabolic Balance

mTOR Signaling in Growth, Metabolism, and Disease

Pancreatic β-Cell Electrical Activity and Insulin Secretion: Of Mice and Men

The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism

AMPK activation can delay aging


AMPK activation/responsiveness decreases with age, resulting in: reduced autophagic clearance of unnecessary products an increase in oxidative stress a decrease resistance to cellular stress


The PI3K/Akt signaling axis in Alzheimer’s disease: a valuable target to stimulate or suppress

Signaling and Metabolism

Increased basal level of Akt-dependent insulin signaling may be responsible for the development of insulin resistance

The PI3K/AKT pathway in obesity and type 2 diabetes

Role of PI3K/AKT Pathway in Insulin-Mediated Glucose Uptake

The PI3K/Akt Pathway in Meta-Inflammation

The PI3K/AKT pathway in obesity and type 2 diabetes

Exercise-Induced Autophagy Suppresses Sarcopenia Through Akt/mTOR and Akt/FoxO3a Signal Pathways and AMPK-Mediated Mitochondrial Quality Control

Akt/PKB activation and insulin signaling: a novel insulin signaling pathway in the treatment of type 2 diabetes

Involvement of AMP-activated protein kinase in neuroinflammation and neurodegeneration in the adult and developing brain

Role of PI3K/AKT Pathway in Insulin-Mediated Glucose Uptake


I. KREBS CYCLE/NAD

AMPK activation/responsiveness decreases with age, resulting in: reduced autophagic clearance of unnecessary products an increase in oxidative stress a decrease resistance to cellular stress


AMPK: Mechanisms of Cellular Energy Sensing and Restoration of Metabolic Balance

BACK TO MENU


I. KREBS CYCLE/NAD

KREBS6

1. Krebs Cycle or Citric Acid Cycle

2. Krebs / citric acid cycle | Cellular respiration | Biology | Khan Academy

3. TCA/Citric Acid (Krebs) Cycle DIRTY MEDICINE

4. Electron Transport Chain (Oxidative Phosphorylation)

5. Pyruvate Pathways & Metabolism

5. KREBS CYCLE NINJA NERD

6. Metabolism - Electron Transport Chain: Overview You Tube

KREBS1

7. Krebs Cycle Reimagined: The Emerging Roles of Succinate and Itaconate as Signal Transducers

8. Krebs’ Cycle Intermediates

Effects of Ketone Bodies on Brain Metabolism and Function in Neurodegenerative Diseases

mtDNA Heteroplasmy at the Core of Aging-Associated Heart Failure. An Integrative View of OXPHOS and Mitochondrial Life Cycle in Cardiac Mitochondrial Physiology

Cervicogenic Headache

Physiology, Krebs Cycle

Mitochondrial pyruvate transport: a historical perspective and future research directions

The role of pyruvate carboxylase in insulin secretion and proliferation in rat pancreatic beta-cells

KREBS1

NADPH and Glutathione Redox Link TCA Cycle Activity to Endoplasmic Reticulum Homeostasis

Citrate – new functions for an old metabolite

NAD

Meet CD38

Different NAD Levels by Age:

BACK TO MENU


ELECTRIC TRANSPORT CHAIN

VIDEO ELECTRON TRANSPORT

VIDEO ATP: The Fuel of Life

LOGO

The Role of Mitochondria in the Pathogenesis of Type 2 Diabetes

Electron Transport Chain (Oxidative Phosphorylation)

BACK TO MENU


BETA OXIDATION

OPEN TEXT

Fatty Acid beta-Oxidation

Randle Cycle

Regulation of Hexokinase Binding to VDAC

The Cori Cycle

Lipid Metabolism

Altered Insulin Signaling in Alzheimer’s Disease Brain – Special Emphasis on PI3K-Akt Pathway

IRS1/PI3K/AKT pathway signal involved in the regulation of glycolipid metabolic abnormalities

VIDEO NN - Metabolism | Fatty Acid Oxidation: Part 1 - IRS1/PI3K/AKT pathway

VIDEO NN - Metabolism | Fatty Acid Oxidation: Part 2

VIDEO NN - Metabolism | The Metabolic Map: Proteins

VIDEO NN - Metabolism | Amino Acid Metabolism

Oxidation of Fatty Acids

BACK TO MENU


NAD+


NAD+

NAD+ in Brain Aging and Neurodegenerative Disorders

BACK TO MENU


ENDOCRINE HORMONES HPA

OPEN TEXT

Hypothalamus-adipose tissue crosstalk: neuropeptide Y and the regulation of energy metabolism

New Insights into the Role of Insulin and Hypothalamic-Pituitary-Adrenal (HPA) Axis in the Metabolic Syndrome

Effect of circadian clock disruption on type 2 diabetes

Thyroid Dysfunction and Diabetes Mellitus: Two Closely Associated Disorders

The Endocrine System

The Endocrinology of Ageing

Effect of circadian clock disruption on type 2 diabetes

Hyperinsulinemia Associated Depression

BACK TO MENU


GLYCOLIPID TOXICITY

GLYCOLIPID1

Biochemical and major pathways underlying endothelial dysregulation in vascular complications of diabetes. Long-term hyperglycemia and hyperlipidemia can cause endothelial cell dysfunction and increase the adhesion of monocytes and platelets. The former can transform into macrophages, while the latter can recruit blood cells, accumulate in blood vessels, and form thrombi. On the one hand, macrophages invade endothelial cells and engulf ox-LDL, turning into foam cells and forming arterial plaques. Macrophages release inflammatory and transcription factors that aggravate the inflammatory response. Excessive glucose and lipid levels will covalently combine to form AGEs, which can bind to their receptors; activate the MAPK and NF-KB pathways, among others; and reduce the production and utilization of NO. Abnormal glucose and lipid metabolism can also affect mitochondrial function, produce excessive ROS, and lead to insufficient energy supply. Epigenetic modifications are also closely related to vascular injury in the vascular complications of T2D, including histone and DNA modifications, and ncRNA regulation.

The mechanisms of glycolipid metabolism disorder on vascular injury in type 2 diabetes

Role of advanced glycation end products on vascular smooth muscle cells under diabetic atherosclerosis

Cardiac Glucolipotoxicity and Cardiovascular Outcomes

Lipotoxicity plays a key role in the development of both insulin resistance and muscle atrophy in patients with type 2 diabetes

The role of fatty acids in insulin resistance

Fatty acid metabolism, energy expenditure and insulin resistance in muscle

Glucolipotoxicity promotes the capacity of the glycerolipid/NEFA cycle supporting the secretory response of pancreatic beta cells

The Randle cycle, the precarious linkbetween sugars and fats

Mechanisms underlying skeletal muscle insulin resistance induced by fatty acids: importance of the mitochondrial function

Trapped fat: Obesity pathogenesis as an intrinsic disorder in metabolic fuel partitioning

ADIPOKINES1

ADIPOKINES2

Chronic inflammation in fat and IR

VISCERAL FAT INFLAMATION

A nexus of lipid and O-Glcnac metabolism in physiology and disease

Integrating Mechanisms for Insulin Resistance: Common Threads and Missing Links

Adipose Morphology: a Critical Factor in Regulation of Human Metabolic Diseases and Adipose Tissue Dysfunction

It Is Not Just Fat: Dissecting the Heterogeneity of Adipose Tissue Function

Adipose tissue plasticity: how fat depots respond differently to pathophysiological cues

BACK TO MENU


LIPOTOXICITY

BACK TO MENU


fat2

Increased tissue (muscle and liver) and plasma fat content, i.e., lipotoxicity, plays a central role in the pathogenesis of type 2 diabetes (1,4,8,27,32–34). Elevated bioactive lipids in the circulation, including lipoproteins, triglycerides, and fatty acids (27), and excessive tissue lipid deposits of long-chain fatty acyl CoAs, diacylglycerol, and ceramide (35,36) have been implicated in the phenomenon of lipotoxicity. Much evidence supports a role for circulating free fatty acids in the development of insulin resistance, inflammation, and β-cell dysfunction (1,3,4,33). Recently, elevated plasma sphingolipids have been implicated in the pathogenesis of obesity-induced cardiovascular and metabolic disease (37). Sphingolipid and ceramide formation are stimulated by inflammatory cytokines, such as TNF-α, which is released from adipocytes and elevated in the plasma of type 2 diabetic and obese subjects (5,6). REFER TO THE FIRST REFERENCE BELOW.

Plasma Ceramides Are Elevated in Obese Subjects With Type 2 Diabetes and Correlate With the Severity of Insulin Resistance

Emerging Roles of Ceramide in Cardiovascular Diseases

AMP kinase and malonyl-CoA: Targets for therapy of the metabolic syndrome

Aging adipose: Depot location dictates age-associated expansion and dysfunction

The Transport of Fatty Acids: Roles of Adipose Tissue Proteins


BACK TO MENU




FRUCTOSE

FRUC19

A comparison of the hepatic fructose (left) and glucose (right) metabolism after consumption of high loads of sugar in the form of SSB. It is hypothesized that an increased de novo lipogenesis after fructose intake in parallel with a decreased fatty acid oxidation leads to hepatic fat deposition. ACC, acetyl-CoA-carboxylase; ATP, adenosine triphosphate; CPT1a, carnitine palmitoyltransferase 1A; FA, fatty acid; GLUT, glucose transporter; KHK-C, ketohexokinase-C; Ox, oxidation; P, phosphate; SSB, sugar-sweetened beverage; TCA, tricarboxylic acid cycle. the follow will try to explain the pathophysiology of fructose, considering overnutrition, gluconeogenesis, regulation, etc.. Fructose is a simple sugar (monosaccharide) found in Sucrose, ultra-processed foods, fruits, honey, and high-fructose corn syrup (HFCS). Its metabolism and pathophysiology differ significantly from glucose, particularly in the context of overnutrition and metabolic regulation. Below is an explanation of the pathophysiology of fructose, focusing on its metabolism, effects on gluconeogenesis, and regulatory mechanisms, especially in the setting of excessive intake.
______________________________________
_

_
1. Fructose Metabolism
ºFructose is primarily metabolized in the liver, though some metabolism occurs in the intestines and kidneys. The key steps in fructose metabolism are:
style="border: 3px solid red;">º Absorption: Fructose is absorbed in the small intestine via the GLUT5 transporter.
º Hepatic metabolism: In the liver, fructose is rapidly phosphorylated by fructokinase to form fructose-1-phosphate (F1P).
This reaction bypasses the rate-limiting step of glycolysis (phosphofructokinase-1), allowing fructose to enter metabolic pathways more rapidly than glucose.
º Cleavage: F1P is cleaved by aldolase B into dihydroxyacetone phosphate (DHAP) and glyceraldehyde, which can enter glycolysis or gluconeogenesis.
2. Effects of Overnutrition
º Excessive fructose consumption, particularly from added sugars like HFCS, has been linked to metabolic dysregulation. Key pathophysiological effects include:
º Lipogenesis and Fatty Liver
º Fructose metabolism generates substrates (e.g., acetyl-CoA) that promote de novo lipogenesis (DNL), leading to triglyceride synthesis.
º This contributes to non-alcoholic fatty liver disease (NAFLD) and hepatic insulin resistance.
º Fructose does not stimulate insulin secretion directly, but chronic overconsumption leads to:
º Increased hepatic glucose production.
º Impaired insulin signaling in peripheral tissues.
º Systemic insulin resistance, a hallmark of metabolic syndrome.
º Uric Acid Production
º Fructose metabolism depletes ATP, leading to increased production of uric acid as a byproduct.
º Elevated uric acid levels are associated with hypertension, inflammation, and gout.
º Appetite Dysregulation
º Fructose does not stimulate leptin (satiety hormone) or suppress ghrelin (hunger hormone) as effectively as glucose.
This can lead to overeating and weight gain.
3. Gluconeogenesis and Fructose
º Fructose can serve as a substrate for gluconeogenesis, the process by which the liver produces glucose from non-carbohydrate sources. However, excessive fructose intake has paradoxical effects:
º Increased gluconeogenesis: Fructose provides carbons for glucose synthesis, but overnutrition can lead to excessive glucose production, contributing to hyperglycemia.
º Dysregulation: Chronic fructose consumption impairs the normal regulation of gluconeogenesis, exacerbating insulin resistance and metabolic dysfunction.
4. Regulation of Fructose Metabolism
ºFructose metabolism is less tightly regulated than glucose metabolism, which contributes to its pathophysiological effects:
º Fructokinase: This enzyme is not regulated by feedback inhibition, allowing uncontrolled entry of fructose into metabolic pathways.
º Hormonal regulation: Unlike glucose, fructose metabolism is not directly regulated by insulin. However, insulin resistance induced by fructose can indirectly affect its metabolism.
º AMP deaminase activation: Fructose metabolism depletes ATP, activating AMP deaminase and increasing uric acid production, which further exacerbates metabolic stres
5. Long-Term Consequences of Excessive Fructose Intake
ºChronic overconsumption of fructose is associated with:
º Obesity: Due to its effects on appetite regulation and lipogenesis.
º Type 2 diabetes: Driven by insulin resistance and impaired glucose homeostasis.
º Cardiovascular disease: Linked to dyslipidemia, hypertension, and inflammation.
º NAFLD: Resulting from excessive hepatic lipid accumulation.
6. Summary Fructose metabolism, while efficient, becomes pathophysiological in the context of overnutrition. Its unregulated entry into metabolic pathways, promotion of lipogenesis, and contribution to insulin resistance and gluconeogenesis dysregulation underlie its role in metabolic syndrome and related disorders. Reducing excessive fructose intake, particularly from added sugars, is crucial for preventing these adverse health outcomes.


FRUC17


Refer to PMC10488931 FOR DETAILS and (referances). Main pathways of fructose metabolism by the liver. For the sake of clarity, only fructose reactions are shown in this figure, on the understanding that they occur at the same time as glycolysis. The importance of the interactions between glucose and fructose metabolism in the liver is explained in the text, as well as in Figure 3 and Figure 4. More than 90% of absorbed fructose is retained in the liver. Fructose from the portal vein enters the hepatocyte (1) via transporter Glut 2 (a high-capacity transporter also used by glucose) and, secondarily, by Glut 8. The phosphorylation of fructose requires a specific enzyme, either fructokinase or ketohexoquinase C (KHK C) (2). This phosphorylation is very quick and is unregulated (unlike glucose phosphorylation by glucokinase) and may lead to the depletion of ATP if the concentration of fructose is very high (3). Ultimately, the recycling of ADP leads to the production of AMP (4), which is then transformed into uric acid (5). Another enzyme that is specific for the metabolism of fructose is aldolase B (6), which leads to the production of the classic trioses of glycolysis (7) with the aid of triokinase (8). An increase in the flux of trioses may lead, under certain circumstances, to the production of methylglyoxal (9) and advanced glycation end products (10) that damage proteins, lipids, and DNA. Since, usually, fructose is ingested at the same time and in the same amounts as glucose, which is converted largely into glycogen or sent to the circulation, the extra trioses are mostly shunted to acetyl-coA (11) and then enter the pathway for the synthesis of fatty acids, also known as de novo lipogenesis (DNL), via malonyl-coA (12). This molecule commits these carbons to fat as it inhibits CPT1 (13), hindering FA entry into the mitochondria for oxidation. Either by providing the carbons as shown in the figure, or by stimulus (ChREBP and SREBP1c), as discussed in the text and in Figure 3 and Figure 4, fructose provides the trioses for the backbone of triglycerides (TG) and the fatty acids as well (14). TG may accumulate as liver fat (15), or be secreted as VLDL (16) or both, depending on fluxes, other substrates, genetics, and hormones. Overactive DNL, caused by a constant excess of fructose, also leads to the production of ceramides, which may be the basis of hepatic insulin resistance, further complicating the metabolic derangement (17). This figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.

FRUC18


Refer to PMC10488931 FOR DETAILS and (referances). Crosstalk between fructose and glucose metabolism in the liver: the role of F-1-P as a signaling molecule of abundance that has gone awry in current dietary habits. The ingestion of fructose is usually accompanied by a similar amount of glucose, be it in the form of sugar or high-fructose corn syrup. Akin to fructose, glucose is also taken up by Glut 2 transporters in the hepatocytes (1). Fructose-1-P (2), the product of fructokinase, acts as a signaling molecule that activates glucose metabolism by its action on glucokinase, GK (3), and pyruvate kinase (4), resulting in an increased flux of intermediates. After the saturation of glycogenesis (5), these lead to DNL (6) and esterification, with TG as end products (7). TG are packed with apoB100 in VLDL (8) and/or stored as liver fat, as shown in Figure 2. Fructose-1-P induces the expression of hypoxia-inducible factor 1 subunit alpha (HIF1a) (9), which, in turn, promotes microvilli expansion and increased absorptive surface (10), thereby enhancing energy uptake and TG production. This figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.



Fructose is a ketohexose and sweetest among all the natural sugars. Like other reducing sugars, it reacts readily with the amino- and nucleophilic groups of proteins, nucleic acids and other biomolecules resulting in glycation reactions. The non-enzymatic glycation reactions comprise Schiff base formation, their Amadori rearrangement followed by complex and partly incompletely understood reactions culminating in the formation of Advance Glycation End products (AGEs). The AGEs are implicated in complications associated with diabetes, cardiovascular disorders, Parkinson's disease, etc. Fructose is highly reactive and forms glycation products that differ both in structure and reactivity as compared to those formed from glucose. Nearly all tissues of higher organisms utilize fructose but only a few like the ocular lens, peripheral nerves erythrocytes and testis have polyol pathway active for the synthesis of fructose. Fructose levels rarely exceed those of glucose but, in tissues that operate the polyol pathway, its concentration may rise remarkably during diabetes and related disorders. Diet contributes significantly to the body fructose levels however, availability of technologies for the large scale and inexpensive production of fructose, popularity of high fructose syrups as well as the promotion of vegetarianism have resulted in a remarkable increase in the consumption of fructose. In vivo glycation reactions by fructose, therefore, assume remarkable significance. The review, therefore, aims to highlight the uniqueness of glycation reactions with fructose and its role in some pathophysiological situations.


Fructose Metabolism (Fructolysis): Steps and Importance

Effects of Fructose vs Glucose on Regional Cerebral Blood Flow in Brain Regions Involved With Appetite and Reward Pathways

Formation of Fructose-Mediated Advanced Glycation End Products and Their Roles in Metabolic and Inflammatory Diseases

Hypothesis: Could Excessive Fructose Intake and Uric Acid Cause Type 2 Diabetes?

Biochemistry, Fructose Metabolism

Fructose metabolism and metabolic disease

What’s the Difference Between Fructose and Glucose?

Molecular aspects of fructose metabolism and metabolic disease

Alcoholism and Diabetes Mellitus

Sugar consumption, metabolic disease and obesity: The state of the controversy

Sugar, Uric Acid, and the Etiology of Diabetes and Obesity

Biochemistry, Polyol Or Sorbitol Pathways

POLYOL PATHWAY

Isocaloric fructose restriction and metabolic improvement in children with obesity and metabolic syndrome

Unifying mechanism for diabetic complications

Fructose Metabolism Biochemistry Topics, Carbohydrate Metabolism

Regulation of the fructose transporter GLUT5 in health and disease

Fructose Metabolism and Cardiac Metabolic Stress

Dietary fructose as a metabolic risk factor

Biochemistry, Fructose Metabolism

The sweet path to metabolic demise: fructose and lipid synthesis

Fructose metabolism and metabolic disease

Formation of Fructose-Mediated Advanced Glycation End Products and Their Roles in Metabolic and Inflammatory Diseases1,2

Fructose Metabolism (Fructolysis): Steps and Importance

Deletion of fructokinase in the liver or in the intestine reveals differential effects on sugar-induced metabolic dysfunction

Fructose Metabolism (Fructolysis): Steps and Importance

Fructose Metabolism (Fructolysis): Steps and Importance

Fructose: A Key Factor in the Development of Metabolic Syndrome and Hypertension

VIDEO: The Dangers of Fructose & Uric Acid - with Dr. Richard Johnson | The Empowering Neurologist

Metabolic Stability of D-Allulose in Biorelevant Media and Hepatocytes: Comparison with Fructose and Erythritol

Serum glucose and insulin levels and erythritol balance after oral administration of erythritol in healthy subjects

Erythritol Attenuates Postprandial Blood Glucose by Inhibiting α-Glucosidase

D-ALLOUSE Comparison with Fructose and Erythritol

Fructose Impact on Brain Function in Rodent Models

Fructose metabolism and metabolic disease

Endogenous fructose production: what do we know and how relevant is it?

Energy and Fructose From Beverages Sweetened With Sugar or High-Fructose Corn Syrup Pose a Health Risk for Some People1

Fructose Malabsorption and Intolerance: Effects of Fructose with and without Simultaneous Glucose Ingestion

High Fat and High Sucrose (Western) Diet Induce Steatohepatitis that is Dependent on Fructokinase

Uric Acid Stimulates Fructokinase and Accelerates Fructose Metabolism in the Development of Fatty Liver

Perspective: A Historical and Scientific Perspective of Sugar and Its Relation with Obesity and Diabetes

Sugar-sweetened beverages and weight gain in children and adults: a systematic review and meta-analysis

Added Sugar Intake and Cardiovascular Diseases Mortality Among US Adults

Uric acid induces fat accumulation via generation of endoplasmic reticulum stress and SREBP-1c activation in hepatocytes

Fructose and NAFLD: The Multifaceted Aspects of Fructose Metabolism

Added Fructose in Non-Alcoholic Fatty Liver Disease and in Metabolic Syndrome:

Fructose as a metabolic toxin that targets the gut-liver axis

High Fructose Intake and Adipogenesis

Fructose Consumption in the Development of Obesity and the Effects of Different Protocols of Physical Exercise on the Hepatic Metabolism

Fructose, insulin resistance, and metabolic dyslipidemia

Pathophysiology of NAFLD and NASH in Experimental Models: The Role of Food Intake Regulating Peptides

Systems Nutrigenomics Reveals Brain Gene Networks Linking Metabolic and Brain Disorders

Chronic Consumption of Fructose Induces Behavioral Alterations by Increasing Orexin and Dopamine

Fructose impairs fat oxidation: Implications for the mechanism of western diet-induced NAFLD

Fructose and Uric Acid: Major Mediators of Cardiovascular Disease Risk Starting at Pediatric Age

Fructose and NAFLD: The Multifaceted Aspects of Fructose Metabolism

High Fructose Intake and Adipogenesis

Formation of Fructose-Mediated Advanced Glycation End Products and Their Roles in Metabolic and Inflammatory Diseases

ChREBP regulates fructose-induced glucose production independently of insulin signaling

Testing the carbohydrate-insulin model in mice:

Dietary fructose enhances tumour growth indirectly via interorgan lipid transfer

Fructose Hijacks the Liver to Grow Cancer

Fructose contributes to the Warburg effect for cancer growth

Dietary Fructose: Implications for Dysregulation of Energy Homeostasis and Lipid/Carbohydrate Metabolism

Endocrine and metabolic effects of consuming beverages sweetened with fructose, glucose, sucrose, or high fructose corn syrup

Recent Progress on Fructose Metabolism—Chrebp, Fructolysis, and Polyol Pathway

Dietary Fructose and Fructose-Induced Pathologies

Consumption of Sugar-Sweetened Beverages Has a Dose-Dependent Effect on the Risk of Non-Alcoholic Fatty Liver Disease:

Fructose drives de novo lipogenesis affecting metabolic health

Sugar Making Sugar: Gluconeogenesis Triggered by Fructose via a Hypothalamic-Adrenal-Corticosterone Circuit

Sugar and Dyslipidemia: A Double-Hit, Perfect Storm

Role of Dietary Fructose and Hepatic de novo Lipogenesis in Fatty Liver Disease

Sweet but Bitter: Focus on Fructose Impact on Brain Function in Rodent Models

ChREBP regulates fructose-induced glucose production independently of insulin signaling

The intestinal-hepatic axis: a comprehensive review on fructose metabolism and its association with mortality and chronic metabolic diseases

Fructose and Sugar: A Major Mediator of Nonalcoholic Fatty Liver Disease

Role of Dietary Fructose and Hepatic de novo Lipogenesis in Fatty Liver Disease

Dietary Fructose and the Metabolic Syndrome



A short review on the aetiology and pathophysiology of alcoholism

Alcohol and Metabolic-associated Fatty Liver Disease


BACK TO MENU




GLP-1


GLP-1-10

Circumventing the issue of cell specificity, is the exploitation of glucagon-like-peptide-1 (GLP-1), as this interacts with a receptor that is only expressed on β-cells (and neurons). GLP-1 is a gut-derived incretin that is naturally produced in response to food ingestion to enhance insulin secretion. Specifically, GLP-1 has been reported to promote proliferation and inhibit apoptosis of β-cells, in vivo and in vitro, and to attenuate apoptosis induced by several pro-apoptotic agents (such as STZ and reactive oxygen species [ROS]).35, 36 Chang et al (2016) showed that GLP-1 suppressed methylglyoxal (MG)-induced apoptosis in a rat insulinoma cell line (RINm5F) by improving mitochondrial function and activating PKA and PI3K/Akt signaling, thereby causing Akt phosphorylation and subsequent inhibition of the cleavage (ie, activation) of pro-apoptotic caspase-3. These effects were corroborated by experiments using MIN6 and INS-1 β-cells.37 In addition to enhancing proliferation and survival, GLP-1 has been recently shown to putatively promote β-cell neogenesis in a severe insulin-deficient diabetic rat model induced by administration of a single high dose of STZ. Indeed, the consequent increase in β-cell number following GLP-1 treatment was not attributable to β-cell replication but rather to α-cell dedifferentiation and subsequent transdifferentiation into glucose-responsive insulin-secreting β-cells. This was associated with the regulation of the GLP-1 receptor and its downstream transcription factor pathway PI3K/Akt/FOXO1.35


GLP6

While these beneficial effects of GLP-1 strongly support its use as a therapeutic agent, it is rapidly degraded in vivo (t1/2 ~ 2 minutes) by the endogenous enzyme dipeptidyl-peptidase-IV (DPP-4).38 To extend bioavailability to improve clinical efficacy, several synthetic GLP-1 receptor agonists have been developed. For instance, the native molecule exendin-4 has been shown to enhance β-cell proliferation and inhibit apoptosis, both in vitro and in vivo, thereby increasing β-cell number and mass, respectively. These effects were abolished in the presence of the PI3K inhibitor LY294002, establishing that the pro-survival/proliferation effect was mediated by PI3K/Akt signaling in β-cells.39 Likewise, the human GLP-1 analogue liraglutide has been shown to promote β-cell proliferation and inhibit apoptosis, both in vitro and in vivo. Indeed, in BTC-6 β-cells, liraglutide suppressed apoptosis induced by serum withdrawal through PI3K/Akt phosphorylation, leading to the inhibition of caspase-3 activity via a mechanism like that of GLP-1. The downstream Akt targets, pro-apoptotic Bad and FOXO1 transcription factor, also underwent inhibitory phosphorylation. These observations were corroborated in an animal model of overt diabetes where liraglutide restored islet size, ameliorated β-cell apoptosis, and increased expression levels of nephrin, 40 a key protein involved in β-cell survival signaling .41 Despite the positive effects on β-cell survival in vivo, there are some concerns regarding the safety profile of these GLP-1 analogues due to their ability to enhance the proliferation of cells. While a consensus has not been reached, evidence from some clinical studies has shown an expansion of exocrine and endocrine pancreatic cells with a possible association to pancreatic cancer.42

As an alternative biologic, γ-aminobutyric acid (GABA) may be a candidate therapeutic agent. It is endogenously produced in β-cells and has been found to stimulate β-cell proliferation in mouse and human islets through the PI3K/mTORC1 pathway. Co-treatment of mice with both GABA and Ly49, a novel GABA type A (GABAA) receptor-positive allosteric modulator, amplified these positive effects. Furthermore, co-treatment with GABA and Ly49 increased β-cell area and proliferation, as compared to mice treated with GABA alone, and similar observations were made using human islets.43 Importantly, a number of GABAA receptor agonists are currently in clinical use as a treatment for epilepsy,44 which suggests that they are considered safe for use in humans and so could potentially be tested in clinical trials as a treatment for T1D.



Targeting the PI3K/Akt signaling pathway in pancreatic β-cells to enhance their survival and function

In Alzheimer’s Disease, 6-Month Treatment with GLP-1 Analog Prevents Decline of Brain Glucose Metabolism: Randomized, Placebo-Controlled, Double-Blind Clinical Trial

Glucagon-like peptide 1-based therapies for the treatment of type 2 diabetes mellitus

GLP-1 Receptor Agonist as Adjuvant Therapy in Type 1 Diabetes: No Apparent Benefit for Beta-Cell Function or Glycemia

Hyperglycemia Potentiates the Slowing of Gastric Emptying Induced by Exogenous GLP-1

Hyperglycemia Potentiates the Slowing of Gastric Emptying Induced by Exogenous GLP-1

GLP-1 receptor activated insulin secretion from pancreatic β-cells: mechanism and glucose dependence

Diabetes drugs in the GLP-1 agonists

MAYO CLINIC GLP-1 Agonists

Glucagon-like peptide 1 (GLP-1)

The Physiology of Glucagon-like Peptide 1

The Capacity to Secrete Insulin Is Dose-Dependent to Extremely High Glucose Concentrations: A Key Role for Adenylyl Cyclase

Potential preventive properties of GLP-1 receptor agonists against prostate cancer:


BACK TO MENU




ENDIOTHELIAL CELL DYSFUNCTION

ENDO1

Endothelial cells become dysfunctional in high glucose environments, which is connected to glucose metabolism and transport. In normal glucose, endothelial cells take up glucose from the blood via glucose transporters (GLUT) and sodium-glucose cotransporters (SGLT). Glucose can then be metabolized, primarily via glycolysis. Pyruvate is shuttled into the mitochondria to be metabolized in the tricarboxylic acid (TCA) cycle or converted to lactate and transported out of the cell via monocarboxylate transporters (MCT). Alternatively, glucose can be transported to the vessel wall or parenchymal tissue paracellularly through cell–cell junctions or transcellularly via GLUTs and SGLTs at the cell abluminal surface. In high glucose, endothelial cells take up more glucose and increase glucose metabolism via glycolysis. Some of the excess glycolytic intermediate metabolites are shunted down glycolytic side branch pathways that contribute to endothelial dysfunction, including the polyol, pentose phosphate, hexosamine biosynthetic, and methylglyoxal pathways. Side branch pathway metabolism is further increased by superoxide overproduction from excess mitochondrial metabolism. Excess glucose also increases protein kinase C (PKC) activation, changes microRNA (miRNA) and long non-coding RNA (lncRNA) expression, and increases extracellular vesicle (EV) release while changing EV contents.



BLOODCELL1

The Relationship between Erythrocytes and Diabetes Mellitus

Vascular actions of insulin with implications for endothelial dysfunction

Endothelial dysfunction and diabetes: roles of hyperglycemia, impaired insulin signaling and obesity

Role of Lipotoxicity in Endothelial Dysfunction

Role of free fatty acids in endothelial dysfunction

A Narrative Review of Diabetic Macroangiopathy: From Molecular Mechanism to Therapeutic Approaches

Endothelial response to glucose: dysfunction, metabolism, and transport

Vascular endothelial dysfunction, a major mediator in diabetic cardiomyopathy

Endothelial dysfunction in diabetes mellitus

Modulation of endothelium function by fatty acids

Endothelial Toxicity of High Glucose and its by-Products in Diabetic Kidney Disease

Physiological and pathological characteristics of vascular endothelial injury in diabetes and the regulatory mechanism of autophagy

Endothelial response to glucose: dysfunction, metabolism, and transport

Nitric oxide decreases cytokine-induced endothelial activation. Nitric oxide selectively reduces endothelial expression of adhesion molecules and proinflammatory cytokines.

Reciprocal Relationships Between Insulin Resistance and Endothelial Dysfunction

Endothelial response to glucose: dysfunction, metabolism, and transport


BACK TO MENU




holding

OPEN

The association between blood glucose levels and lipids or lipid ratios in type 2 diabetes patients

The Time Is Right for a New Classification System for Diabetes: Rationale and Implications of the β-Cell–Centric Classification Schema

Latent autoimmune diabetes in adults (LADA)

Mechanisms of Diabetic Complications

Cognitive decline and dementia in diabetes mellitus: mechanisms and clinical implications

Comprehensive review on the pathogenesis of hypertriglyceridaemia-associated acute pancreatitis

An enzyme that selectively S-nitrosylates proteins to regulate insulin signaling

ADA Differentiation of Diabetes by Pathophysiology, Natural History, and Prognosis

The Ketogenic Effect of Medium-Chain Triacylglycerides

Astrocyte metabolism of the medium-chain fatty acids octanoic acid and decanoic acid promotes GABA synthesis in neurons via elevated glutamine supply

Applications of Medium-Chain Triglycerides in Foods

Identification and Mechanism of 10-Carbon Fatty Acid as Modulating Ligand of Peroxisome Proliferator-activated Receptors

Effect of Different Medium-Chain Triglycerides on Glucose Metabolism in High-Fat-Diet Induced Obese Rats

Apolipoprotein D

Effect of circadian clock disruption on type 2 diabetes

Cas9 screens reveal regulators of ageing in neural stem cells

Combined metabolic activators improve cognitive functions in Alzheimer’s disease patients: a randomised, double-blinded, placebo-controlled phase-II trial

Efficacy and Safety of Berberine Alone for Several Metabolic Disorders: A Systematic Review and Meta-Analysis of Randomized Clinical Trials

VIDEO NINJA KETONES

The genetic architecture of protein stability

Mutations that affect protein stability follow simple math rules, study shows

Endocrine role of bone in the regulation of energy metabolism

A Metabolic Enhancer Protects against Diet-Induced Obesity and Liver Steatosis and Corrects a Pro-Atherogenic Serum Profile in Mice

Exercise-induced exerkines: Secret weapon in disease prevention and therapy

Exercise, exerkines, and cardiometabolic health: from individual players to a team sport

Exerkines’: A Comprehensive Term for the Factors Produced in Response to Exercise

Novel Multi-Ingredient Supplement Facilitates Weight Loss and Improves Body Composition in Overweight and Obese Individuals:

Lee Index

Comprehensive Prognostic Tool for Adults ≥ 70

Obesity and Nonalcoholic Fatty Liver Disease: Biochemical, Metabolic and Clinical Implications


BACK TO MENU




LIVER NAFLD TO DIABETES

Nutrient overload occurs worldwide as a consequence of the modern diet pattern and the physical inactivity that sometimes accompanies it. Cells initiate multiple protective mechanisms to adapt to elevated intracellular metabolites and restore metabolic homeostasis, but irreversible injury to the cells can occur in the event of prolonged nutrient overload. Many studies have advanced the understanding of the different detrimental effects of nutrient overload; however, few reports have made connections and given the full picture of the impact of nutrient overload on cellular metabolism. In this review, detailed changes in metabolic and energy homeostasis caused by chronic nutrient overload, as well as their associations with the development of metabolic disorders, are discussed. Overnutrition-induced changes in key organelles and sensors rewire cellular bioenergetic pathways and facilitate the shift of the metabolic state toward biosynthesis, thereby leading to the onset of various metabolic disorders, which are essentially the downstream manifestations of a misbalanced metabolic equilibrium. Based on these mechanisms, potential therapeutic targets for metabolic disorders and new research directions are proposed.


Overnutrition, particularly a diet high in fructose (from sources like high-fructose corn syrup and sucrose), high glycemic load carbohydrates, and fat should cause a metabolic cascade of:

_
OVERNUTRITION
1. Lipogenesis
2. De Novo Lipolysis:
3. De Novo Gluconeogenesis as Overnutrition should cause regulation if glycolysis:
4. Elevated levels of Palmitate:
5. Hyperglycemia:
6. Hyperinsulinemia:
7. Increased levels of P300 in the liver through the leaky gut.
8. Insulin resistance in the liver directly:
9. Huge Metabolic problems:
10. This should cause cytokines to be circulated IL6 etc
11. etc..


Hepatic Insulin Clearance: Mechanism and Physiology

Obese visceral fat tissue inflammation: from protective to detrimental?

Removal of Visceral Fat Prevents Insulin Resistance and Glucose Intolerance of Aging: An Adipokine-Mediated Process?

Basic concepts in nutrition: Overnutrition – Functional and clinical consequences

Overnutrition causes insulin resistance and metabolic disorder through increased sympathetic nervous system activity

The sympathetic nervous system in the 21st century: Neuroimmune interactions in metabolic homeostasis and obesity

Insulin resistance drives hepatic de novo lipogenesis in nonalcoholic fatty liver disease

Pathophysiology of NAFLD and NASH in Experimental Models: The Role of Food Intake Regulating Peptides

β2-Adrenergic receptor agonist induced hepatic steatosis in mice:

Obesity and Nonalcoholic Fatty Liver Disease: Biochemical, Metabolic and Clinical Implications

Alcohol and Metabolic-associated Fatty Liver Disease

From overnutrition to liver injury: AMP-activated protein kinase in nonalcoholic fatty liver diseases


BACK TO MENU




PANCREAS

OPEN

PAN11


The Brain-Gut-Islet Connection

The Mechanism of Insulin Release by Pancreatic β-cells

Pancreas Structure and Function | Digestive System DM

Exocrine pancreas | Gastrointestinal system physiology | NCLEX-RN | Khan Academy

Bile Synthesis and Function DM

Pancreas Function, Enzymes & Role in Digestion

Regulation of Whole-Organ Pancreatic Secretion

GABA and Glutamate: Their Transmitter Role in the CNS and Pancreatic Islets

Caffeine in hot drinks elicits cephalic phase responses involving cardiac activity

Endocrine Cephalic Phase Responses to Food Cues: A Systematic Review

A critical role for b cell M3 muscarinic acetylcholine receptors in regulating insulin release and blood glucose homeostasis in vivo

Nitric oxide interacts with cholinoceptors to modulate insulin secretion by pancreatic β cells

Pancreatic regulation of glucose homeostasis

Dissecting the Brain/Islet Axis in Metabesity

Cholesterol Redistribution in Pancreatic β-Cells: A Flexible Path to Regulate Insulin Secretion

Nitric oxide interacts with cholinoceptors to modulate insulin secretion by pancreatic β cells

Control of Insulin Secretion by Cholinergic Signaling in the Human Pancreatic Islet

Glucose-sensing mechanisms in pancreatic β-cells

Pancreas-Brain Crosstalk

Pancreatic regulation of glucose homeostasis

Regulation of Pancreatic Secretion

Neural Control of the Pancreas

Pancreas-Brain Crosstalk

Role of metabotropic glutamate receptors in the regulation of pancreatic functions

Correlation between Pancreatic Lipase Levels and Alzheimer’s Disease Progression

The ageing pancreas: a systematic review of the evidence and analysis of the consequences

Pancreas—Its Functions, Disorders, and Physiological Impact on the Mammals’ Organism

Autonomic dysfunction in patients with Alzheimer’s disease

Diagnosis and Management of Autonomic Dysfunction in Dementia Syndromes

Relationships Between the Autonomic Nervous System and the Pancreas Including Regulation of Regeneration and Apoptosis

Vagal neurocircuitry and its influence on gastric motility

The dorsal motor nucleus of the vagus and regulation of pancreatic secretory function

Pancreas-Brain Crosstalk

Pancreatic Insufficiency

Low serum amylase in association with metabolic syndrome and diabetes

VIDEO Insulin Secretion

VIDEO Incretin Effect: Why oral glucose causes more insulin secretion

VIDEO Mechanism of Action of Insulin

VIDEO Insulin synthesis and mechanism of action - 3D Animation

VIDEO The Mechanism of Insulin Release by Pancreatic β-cells

VIDEO The Mechanism of Insulin Biosynthesis by Pancreatic β-cells

Molecular Biology of KATP Channels and Implications for Health and Disease

Loss of mTORC1 signaling alters pancreatic α cell mass and impairs glucagon secretion

Incretin Hormones and Type 2 Diabetes—Mechanistic Insights and Therapeutic Approaches

Improvement of Glucose Tolerance by Food Factors Having Glucagon-Like Peptide-1 Releasing Activity

The T-type calcium channel CaV3.2 regulates insulin secretion in the pancreatic β-cell

Pancreatic regulation of glucose homeostasis

Recent insights into mechanisms of β-cell lipo- and glucolipotoxicity in type 2 diabetes

pancreatic islets impair insulin secretion due to dysregulated calcium dynamics, glucose responsiveness and mitochondrial activity

Recent Insights Into Mechanisms of β-Cell Lipo- and Glucolipotoxicity in Type 2 Diabetes

Cystine/glutamate antiporter System xc- deficiency impairs insulin secretion in mice

β‐Cell glutamate signaling: Its role in incretin‐induced insulin secretion

An excessive increase in glutamate contributes to glucose-toxicity in β-cells via activation of pancreatic NMDA receptors in rodent diabetes

Role of incretin hormones in the regulation of insulin secretion in diabetic and nondiabetic humans

Role of incretin hormones in the regulation of insulin secretion in diabetic and nondiabetic humans

Glucose‐dependent insulinotropic polypeptide and glucagon‐like peptide‐1: Incretin actions beyond the pancreas

Boosting GLP-1 by Natural Products

Berberine: Ins and outs of a nature-made PCSK9 inhibitor

Sensory nerves and pancreatitis

Alzheimer 's disease and other neurodegenerative diseases may be due to nutritional deficiencies secondary to unrecognized exocrine pancreatic insufficiency

An excessive increase in glutamate contributes to glucose-toxicity in β-cells via activation of pancreatic NMDA receptors in rodent diabetes

Metabolic cycling in control of glucose-stimulated insulin secretion

Glucose‐stimulated insulin secretion: A newer perspective

Mechanisms of the Amplifying Pathway of Insulin Secretion in the β Cell

Long-Term Exposure to Glucose and Lipids Inhibits Glucose-Induced Insulin Secretion Downstream of Granule Fusion With Plasma Membrane

Blocking Ca2+ Channel β3 Subunit Reverses Diabetes

VIDEO: The Birth of Beta Cells

The dorsal motor nucleus of the vagus and regulation of pancreatic secretory function

GLP-1 receptor signalling promotes β-cell glucose metabolism via mTOR-dependent HIF-1α activation

Pancreatic signals controlling food intake; insulin, glucagon and amylin

Glucose-sensing mechanisms in pancreatic β-cells

A pathway model of glucose-stimulated insulin secretion in the pancreatic β-cell

Pancreatic β Cell Mass Death

NADPH Oxidase (NOX) Targeting in Diabetes: A Special Emphasis on Pancreatic β-Cell Dysfunction

Inside the β Cell: Molecular Stress Response Pathways in Diabetes Pathogenesis

The Role of Oxidative Stress in Pancreatic β Cell Dysfunction in Diabetes

β Cell dysfunction during progression of metabolic syndrome to type 2 diabetes

The endoplasmic reticulum stress/autophagy pathway is involved in cholesterol-induced pancreatic β-cell injury

Islet β cell failure in type 2 diabetes

Heterogeneity of Diabetes: β-Cells,

Mitochondrial bioenergetics, metabolism, and beyond in pancreatic β-cells and diabetes

Islet Autoimmunity Is Highly Prevalent and Associated With Diminished β-Cell Function in Patients With Type 2 Diabetes in the GRADE Study

Islet Autoimmunity Is Highly Prevalent and Associated With Diminished β-Cell Function in Patients With Type 2 Diabetes

Impact of Islet Autoimmunity on the Progressive β-Cell Functional Decline in Type 2 Diabetes

Pancreatic Enzyme Secretion

Metformin prevents glucotoxicity by alleviating oxidative and ER stress-induced CD36 expression in pancreatic beta cells

Glucotoxicity promotes aberrant activation and mislocalization of Ras-related C3 botulinum toxin substrate 1 [Rac1] and metabolic dysfunction in pancreatic islet β-cells: Reversal of such metabolic defects by metformin

The Role of CD36 in Type 2 Diabetes Mellitus: β-Cell Dysfunction and Beyond

Insulin Secretion DM

How insulin works

The Mechanism of Insulin Release by Pancreatic β-cells

Production of insulin and glucagon

The Insulinogenic Index Is a Valid Marker of Beta Cell Function in Different Metabolic Categories

Metabolic Signaling in Fuel-Induced Insulin Secretion

Glucose transporters in pancreatic islets

Hypertonicity during a rapid rise in D-glucose mediates first-phase insulin secretion

The role of GLUT2 in glucose metabolism in multiple organs and tissues

Signals and Pools Underlying Biphasic Insulin Secretion

Pancreatic regulation of glucose homeostasis

Age-Related Impairment of Pancreatic Beta-Cell Function: Pathophysiological and Cellular Mechanisms

Mitochondrial Dynamics in the Regulation of Nutrient Utilization and Energy Expenditure

Mitochondrial oxidative stress in aging and healthspan

The pancreatic β-cell in ageing: Implications in age-related diabetes

β cell aging and age-related diabetes

Aging compromises human islet beta cell function and identity by decreasing transcription factor activity and inducing ER stress

Acceleration of β Cell Aging Determines Diabetes and Senolysis Improves Disease Outcomes

Functional changes in beta cells during ageing and senescence

Young capillary vessels rejuvenate aged pancreatic

Pancreatic β-cell senescence in diabetes: mechanisms, markers and therapies

The pancreatic β-cell in ageing: Implications in age-related diabetes

Pancreatic Beta-cell Dysfunction in Type 2 Diabetes

The physiological role of β-cell heterogeneity in pancreatic islet function

The Difference δ-Cells Make in Glucose Control

Pancreatic α and β cells are globally phase-locked

The physiological role of β-cell heterogeneity in pancreatic islet function

Pancreatic regulation of glucose homeostasis


BACK TO MENU




Insulin and Metabolic Syndrome:

Mechanisms of Insulin Action and Insulin Resistance

Metabolic flexibility and insulin resistance

Crosstalk of Mitochondria With Brain Insulin and Leptin Signaling

MECHANISMS OF INSULIN ACTION AND INSULINRESIST

VIDEO: Diabetes and Insulin: A Triumph for Recombinant DNA Technology

Metabolic actions of insulin in men and women

Biochemistry, Insulin Metabolic Effects

Insulin: The master regulator of glucose metabolism

Insulin, insulin-degrading enzyme and amyloid-beta peptide in Alzheimes disease: review and hypothesis

Insulin-degrading enzyme regulates the levels of insulin, amyloid β-protein, and the β-amyloid precursor protein intracellular domain in vivo

Glucose‐stimulated insulin secretion: A newer perspective

glucose‐stimulated insulin release

The key steps leading to glucose-stimulated insulin secretion

The manifold roles of protein S-nitrosylation in the life of insulin

Regulatory role for the arginine-nitric oxide pathway in metabolism of energy substrates

Insulin’s direct hepatic effect explains the inhibition of glucose production caused by insulin secretion

Insulin resistance and Alzheimer’s disease

Gymnema sylvestre: A Memoir

VIDEO: Diabetes and Insulin: A Triumph for Recombinant DNA Technology

Pathogenesis and Pathophysiology of the Cardiometabolic Syndrome

Association between insulin resistance and the development of cardiovascular disease

Insulin Resistance: From Mechanisms to Therapeutic Strategies

Trends in insulin resistance: insights into mechanisms and therapeutic strategy

Mechanisms of Insulin Action and Insulin Resistance

Metabolic flexibility and insulin resistance

The Insulinogenic Index Is a Valid Marker of Beta Cell Function in Different Metabolic Categories

Differential Associations of Oral Glucose Tolerance Test–Derived Measures of Insulin Sensitivity and Pancreatic β-Cell Function With Coronary Artery Calcification and Microalbuminuria in Type 2 Diabetes

Evidence for cephalic phase insulin release in humans: A systematic review and meta-analysis

Use of c-peptide as a measure of cephalic phase insulin release in humans

Metabolic Signaling in Fuel-Induced Insulin Secretion

Crosstalk of Mitochondria With Brain Insulin and Leptin Signaling

1. Introduction

2. Characteristics of glucose transporters

3. Role of PI3K/AKT Pathway in Insulin-Mediated Glucose Uptake

The dynamic clustering of insulin receptor underlies its signaling and is disrupted in insulin resistance

Insulin Receptor Tyrosine Kinase (RTK) - How insulin works

Changes in Cells Associated with Insulin Resistance

D43. Type 2 Diabetes Mellitus and Alzheimer's Disease: Role of Insulin Signalling and Therapeutic Implications

D44. Insulin resistance and Alzheimer’s disease

D46. Insulin-Degrading Enzyme as a Downstream Target of Insulin Receptor Signaling Cascade: Implications for Alzheimer 's Disease Intervention

D40. Insulin resistance and Alzheimer's disease

D37. 17β-Estradiol regulates insulin-degrading enzyme expression via an ERβ/PI3-K pathway in hippocampus: relevance to Alzheimer’s prevention

A critical role for IGF-II in memory consolidation and enhancement

Effects of Regular and Long-Acting Insulin on Cognition and Alzheimer’s Disease Biomarkers: A Pilot Clinical Trial

Alzheimer’s disease and insulin resistance: translating basic science into clinical applications

Insulin dysfunction and Tau pathology

The insulin resistance by triglyceride glucose index and risk for dementia

The dynamic clustering of insulin receptor underlies its signaling and is disrupted in insulin resistance


BACK TO MENU



GLYCATION/AGES

OPEN

The role of glycation in the pathogenesis of aging and its prevention through herbal products and physical exercise

Significance of HbA1c Test in Diagnosis and Prognosis of Diabetic Patients

Glycation and a Spark of ALEs (Advanced Lipoxidation End Products) – Igniting RAGE/Diaphanous-1 and Cardiometabolic Disease

Oral Glycotoxins Determine the Effects of Calorie Restriction on Oxidant Stress, Age-Related Diseases, and Lifespan

Glycation Damage: A Possible Hub for Major Pathophysiological Disorders and Aging

Accumulation of Advanced Glycation End-Products in the Body and Dietary Habits

Advanced Glycation End Products and Diabetic Complications

Role of Glycated Proteins in the Diagnosis and Management of Diabetes: Research Gaps and Future Directions

Analysis of the Binding of Warfarin to Glyoxal- and Methylglyoxal-Modified Human Serum Albumin by Ultrafast Affinity Extraction

Glycated Serum Protein Genetics and Pleiotropy with Cardiometabolic Risk Factors

Methylglyoxal, a highly reactive dicarbonyl compound, as a threat for blood brain barrier integrity

Dicarbonyl stress in clinical obesity

Methylglyoxal-Glyoxalase 1 Balance: The Root of Vascular Damage

Review: Glycation of human serum albumin

Studies of Binding by Sulfonylureas with Glyoxal- and Methylglyoxal-Modified Albumin by Immunoextraction using Affinity Microcolumns

Formation of Fructose-Mediated Advanced Glycation End Products and Their Roles in Metabolic and Inflammatory Diseases

Dietary Sugars and Endogenous Formation of Advanced Glycation Endproducts: Emerging Mechanisms of Disease

Advanced Glycation End Products and Diabetes Mellitus: Mechanisms and Perspectives

The role of advanced glycation end-products in the development of coronary artery disease in patients with and without diabetes mellitus:

Pathological Significance of Mitochondrial Glycation

Glycation Damage: A Possible Hub for Major Pathophysiological Disorders and Aging

Fructose consumption: potential mechanisms for its effects to increase visceral adiposity and induce dyslipidemia and insulin resistance

Uric Acid Stimulates Fructokinase and Accelerates Fructose Metabolism in the Development of Fatty Liver

Sympathetic overactivity precedes metabolic dysfunction in a fructose model of glucose intolerance in mice

Glycation With Fructose:

Prevention of non-enzymatic glycosylation (glycation): Implication in the treatment of diabetic complication

Glycated hemoglobin

Glycated haemoglobin (HbA1c) for the diagnosis of diabetes

Glycation Damage: A Possible Hub for Major Pathophysiological Disorders and Aging

Extracellular superoxide released from mitochondria mediates mast cell death by advanced glycation end products

A Role for Advanced Glycation End Products in Molecular Ageing

Enhanced oxidative stress and damage in glycated erythrocytes

The role of protein kinase C in diabetic microvascular complications

LDL biochemical modifications: a link between atherosclerosis and aging

NADPH Oxidases, Reactive Oxygen Species, and Hypertension

The manifold roles of protein S-nitrosylation in the life of insulin

The AGE–RAGE Pathway and Its Relation to Cardiovascular Disease in Patients with Chronic Kidney Disease

Advanced glycation end products: Key mediator and therapeutic target of cardiovascular complications in diabetes

ABSTRACT: Advanced glycation end products and vascular inflammation: implications for accelerated atherosclerosis in diabetes

Advanced glycation end products: Key mediator and therapeutic target of cardiovascular complications in diabetes

ABSTRACT: Advanced glycation end products and vascular inflammation: implications for accelerated atherosclerosis in diabetes

Effectiveness of Early Advanced Glycation End Product Accumulation Testing in the Diagnosis of Diabetes: A Health Risk Factor Analysis Using the Body Mass Index as a Moderator

Diabetes and Advanced Glycoxidation End Products

Advanced Glycation End Products in Foods and a Practical Guide to Their Reduction in the Diet

The clinical relevance of assessing advanced glycation endproducts accumulation in diabetes

Advanced glycation end products and diabetes and other metabolic indicators

Advanced glycation end products and vascular inflammation: implications for accelerated atherosclerosis in diabetes

Hypoxia Driven Glycation: Mechanisms and Therapeutic Opportunities

Glycosylation Biomarkers Associated with Age-Related Diseases and Current Methods for Glycan Analysis

Advanced Glycation End Products and Risks for Chronic Diseases: Intervening Through Lifestyle Modification

Advanced Glycation End Products (AGEs) IN AGING

Formation of Fructose-Mediated Advanced Glycation End Products and Their Roles in Metabolic and Inflammatory Diseases

Advanced Glycation End-Products and Their Effects on Gut Health

Advanced glycation end products (AGEs) and other adducts in aging-related diseases and alcohol-mediated tissue injury

Dietary Advanced Glycation End Products: Digestion, Metabolism and Modulation of Gut Microbial Ecology

Dietary Advanced Glycation End Products: Their Role in the Insulin Resistance of Aging

Endogenous advanced glycation end products in the pathogenesis of chronic diabetic complications

Advanced Glycation End Products and Diabetes Mellitus: Mechanisms and Perspectives

Role of Hyperglycemia-Induced Advanced Glycation End Product (AGE) Accumulation in Atherosclerosis

The Role of Advanced Glycation End Products in Diabetic Vascular Complications

Advanced Glycation End Products (AGEs), Receptor for AGEs, Diabetes, and Bone:

AGEs, rather than hyperglycemia, areresponsible for microvascular complicationsin diabetes: A “glycoxidation-centric” pointof view

Advanced Glycation Endproducts Stimulate Osteoblast Apoptosis Via the MAP Kinase and Cytosolic Apoptotic Pathways

POLYOL PATHWAY: A REVIEW ON A POTENTIAL TARGET FOR THE PREVENTION OF DIABETIC COMPLICATIONS

Dietary Advanced Glycation End Products Shift the Gut Microbiota Composition and Induce Insulin Resistance in Mice

Dysbiosis-Related Advanced Glycation Endproducts and Trimethylamine N-Oxide in Chronic Kidney Disease

Advanced Glycation End Products in Foods

Diabetic vascular diseases: molecular mechanisms and therapeutic strategies

Formation of Fructose-Mediated Advanced Glycation End Products and Their Roles in Metabolic and Inflammatory Diseases

Advanced glycation end products-induced insulin resistance involves repression of skeletal muscle GLUT4 expression

Advanced glycation end products inhibit glucose-stimulated insulin secretion through nitric oxide-dependent inhibition of cytochrome c oxidase and adenosine triphosphate synthesis

Role for glyoxalase I in Alzheimer's disease

Alcohol-induced gut microbial reorganization and associated overproduction of phenylacetylglutamine promotes cardiovascular disease


BACK TO MENU




OXIDATIVE STRESS

OXS7

OXIDATIVE-STRESS1

Oxidative stress occurs when reactive species are overproduced or when the antioxidant system is weakened. Reaction species are usually derived from oxygen, nitrogen, or sulfur elements, which generate ROS, reactive nitrogen (RNS), and reactive sulfur. ROS and RNS are the major sources of oxidative stress. The free radicals include the superoxide anion radical (O2− ), hydroxyl free radical (OH·), hydrogen peroxide (H2O2) and peroxynitrite (ONOO−).

OXIDATIVE-STRESS2

Reactive oxygen and nitrogen molecules have been typically viewed as the toxic by-products of metabolism. However, accumulating evidence has revealed that reactive species, including hydrogen peroxide, serve as signaling molecules that are involved in the regulation of cellular function. The chronic and/or increased production of these reactive molecules or a reduced capacity for their elimination, termed oxidative stress, can lead to abnormal changes in intracellular signaling and result in chronic inflammation and insulin resistance. Inflammation and oxidative stress have been linked to insulin resistance in vivo. Recent studies have found that this association is not restricted to insulin resistance in type 2 diabetes, but is also evident in obese, nondiabetic individuals, and in those patients with the metabolic syndrome. An increased concentration of reactive molecules triggers the activation of serine/threonine kinase cascades such as c-Jun N-terminal kinase, nuclear factor-kappaB, and others that in turn phosphorylate multiple targets, including the insulin receptor and the insulin receptor substrate (IRS) proteins. Increased serine phosphorylation of IRS reduces its ability to undergo tyrosine phosphorylation and may accelerate the degradation of IRS-1, offering an attractive explanation for the molecular basis of oxidative stress-induced insulin resistance.

Oxidative Stress: Harms and Benefits for Human Health

Oxidative stress signaling in the pathogenesis of diabetic cardiomyopathy and the potential therapeutic role of antioxidant naringenin

Research Progress on Signaling Pathway-Associated Oxidative Stress in Endothelial Cells

Molecular and Chemical Regulation of the Keap1-Nrf2 Signaling Pathway

Resveratrol Inhibits Hydrogen Peroxide-Induced Apoptosis in Endothelial Cells via the Activation of PI3K/Akt by miR-126

Molecular Mechanisms Linking Oxidative Stress and Diabetes Mellitus

OXIDATIVE STRESS, INSULIN SIGNALING AND DIABETES

Lipid Peroxidation: Production, Metabolism, and Signaling Mechanisms of Malondialdehyde and 4-Hydroxy-2-Nonenal

Vagal neurocircuitry and its influence on gastric motility

The etiology of oxidative stress in insulin resistance

Oxidative stress in the pathophysiology of type 2 diabetes and related complications: Current therapeutics strategies and future perspectives

Pathogenesis of Chronic Hyperglycemia: From Reductive Stress to Oxidative Stress

Cellular death, reactive oxygen species (ROS) and diabetic complications

Metabolic Stress, Autophagy and Cardiovascular Aging: from Pathophysiology to Therapeutics

Diabetes and mitochondrial function: Role of hyperglycemia and oxidative stress

Mechanistic Insight into Oxidative Stress-Triggered Signaling Pathways and Type 2 Diabetes

Hyperglycemia and Oxidative Stress: An Integral, Updated and Critical Overview of Their Metabolic Interconnections

Enhanced oxidative stress and damage in glycated erythrocytes

Inside the β Cell: Molecular Stress Response Pathways in Diabetes Pathogenesis

The Role of Oxidative Stress in Pancreatic β Cell Dysfunction in Diabetes

Mitochondrial Dysfunction, Oxidative Stress, and Inter-Organ Miscommunications in T2D Progression

The power of the relaxation response

Genomic Counter-Stress Changes Induced by the Relaxation Response

Relaxation Response Induces Temporal Transcriptome Changes in Energy Metabolism, Insulin Secretion and Inflammatory Pathways

The Role of Oxidative Stress in the Pathogenesis of Diabetic Vascular Complications

OXS8

OXS9


BACK TO MENU



MITOCHONDRIAL DYSFUNCTION

The main function of mitochondria is ATP synthesis through oxidative phosphorylation in response to metabolic demand [115]. Mitochondria also participate in the production of different metabolites used as precursors of several macromolecules (lipids, proteins, and DNA). In addition, mitochondria play an important role in maintaining ion homeostasis, ROS clearance, the stress response, and serve to integrate multiple signaling pathways [116,117]. An imbalance between energy intake and expenditure in the mitochondria generates mitochondrial dysfunction, a state characterized by a reduced ratio of energy production to respiration [112]. Under these circumstances, nutrient oxidation efficiency is reduced leading to a decreased ratio of ATP synthesis/oxygen consumption, which increases O2− production [118]. In fact, the accumulation of ROS in the mitochondria is one proposed mechanism linking mitochondrial dysfunction to IR [119]. This relationship was corroborated in studies showing decreased mitochondria oxidative capacity in skeletal muscle and impaired lipid metabolism in obese and insulin-resistant individuals compared to healthy controls [120,121,122]. In addition, patients with T2DM have been found to have downregulation of genes involved in oxidative metabolism that are regulated by the peroxisome proliferator-activated receptor γ co-activator 1α (PGC 1α) [123,124] and a diminished phosphocreatine re-synthesis rate, both indicative of impaired mitochondrial function [125] (Figure 3). Moreover, some relatives of T2DM patients have been found to have decreased mitochondrial respiration suggesting that mitochondrial dysfunction may precede T2DM development. It has also been proposed that T2DM development may be a direct consequence of defects in the oxidative phosphorylation system and the electron transport chain (ETC) rather than a decrease in mitochondrial content [126].

Assessing mitochondrial dysfunction in cells

The role of mitochondria in metabolic disease: a special emphasis on the heart dysfunction

Metabolic Health, Mitochondrial Fitness, Physical Activity, and Cancer

Mitochondrial Aging and Age-Related Dysfunction of Mitochondria

The Mitochondrial Basis of Aging

Mitochondrial and metabolic dysfunction in ageing and age-related diseases

Aging: All roads lead to mitochondria

The dynamics of mitochondrial DNA heteroplasmy: implications for human health and disease

Mitochondria in migraine pathophysiology – does epigenetics play a role?

Mitochondrial Dysfunction, Oxidative Stress, and Inter-Organ Miscommunications in T2D Progression

Mitochondrial bioenergetics, metabolism, and beyond in pancreatic β-cells and diabetes

Mitochondrial health quality control: measurements and interpretation in the framework of predictive, preventive, and personalized medicine

A Mitochondrial Health Index Sensitive to Mood and Caregiving Stress

Lipid (per) oxidation in mitochondria: an emerging target in the ageing process?

Diabetes and mitochondrial function: Role of hyperglycemia and oxidative stress

The Unfolded Protein Response

Mitochondrial dysfunction: roles in skeletal muscle atrophy

Mitochondrial Dysfunction and Chronic Disease: Treatment With Natural Supplements

Glutathione and mitochondria


BACK TO MENU



STRESS

Stress increases the activity of the SNS and decreases the activity of the PNS, which increases the levels of E and NE, and decreases the level of ACh. This in turn can increase the level of pro-inflammatory cytokines, such as TNF, IL-1, IL-6, and interferons and decrease the level of anti-inflammatory cytokines, such as IL-10, resulting in a state of inflammation.
Endogenous release of epinephrine after stress as well as exogenous epinephrine infusion are known to result in impaired glucose tolerance. Previous studies of man and animals have demonstrated that this effect of epinephrine results from inhibition of insulin secretion and augmentation of hepatic glucose production. Lack of adequate sleep can lead to many complications. Some of these complications can lead to further difficulty in getting sleep. Lack of sleep causes elevated cortisol, which can result in increased blood sugar, increased blood pressure, cravings for carbohydrates, and sugar, which leads to weight gain and other medical and psychiatric complications. The subjective experience of sleep loss can be distressing, which can exacerbate complications. The following is a non-comprehensive list of complications of sleep loss: Diabetes/insulin resistance Hypertension Obesity Obstructive sleep apnea Vascular disease Stroke Myocardial infarct Depression Anxiety Psychosis


Physiology, Stress Reaction

Stress and autonomic response to sleep deprivation in medical residents: A comparative cross-sectional study

Sleep deprivation and stress: a reciprocal relationship

Sleep Deprivation

Stress and Sleep Disorder

Association Between Inpatient Sleep Loss and Hyperglycemia of Hospitalization

Circadian Clock Desynchronization and Insulin Resistance

Intermittent hypoxia in obstructive sleep apnoea mediates insulin resistance through adipose tissue inflammation

The Impact of Intermittent Hypoxia on Metabolism and Cognition

Carotid body chemoreceptors, sympathetic neural activation, and cardiometabolic disease

Sleep dysregulation in sympathetic-mediated diseases: implications for disease progression

A review on linking stress, depression, and insulin resistance via low-grade chronic inflammation

Catecholamine-induced lipolysis causes mTOR complex dissociation and inhibits glucose uptake in adipocytes

Mechanistic Insight into Oxidative Stress-Triggered Signaling Pathways and Type 2 Diabetes


BACK TO MENU



MEMBRBRANE INTEGRITY

Micronutrients May Be a Unique Weapon Against the Neurotoxic Triad of Excitotoxicity, Oxidative Stress and Neuroinflammation: A Perspective

OPEN

BACK TO MENU



INFLAMATION



INFL1

The mechanisms of glycolipid metabolism disorder on vascular injury in type 2 diabetes

Type 2 Diabetes and its Impact on the Immune System

Association of proinflammatory cytokines and islet resident leucocytes with islet dysfunction in type 2 diabetes

Mechanisms Linking Inflammation to Insulin Resistance

Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance

Metabolic Inflammation and Insulin Resistance in Obesity

Vagus Nerve as Modulator of the Brain–Gut Axis in Psychiatric and Inflammatory Disorders

Intakes and sources of dietary sugars and their association with metabolic and inflammatory markers

Potential Synergies of β-Hydroxybutyrate and Butyrate on the Modulation of Metabolism, Inflammation, Cognition, and General Health

Inflammaging decreases adaptive and innate immune responses in mice and humans

Metabolic Stress and Cardiovascular Disease in Diabetes Mellitus: The role of protein O-GlcNAc Modification

NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology

Inflammation, stress, and diabetes

Metabolic Syndrome Is Associated with Oxidative Stress and Proinflammatory State

Inflammation in diabetes complications: molecular mechanisms and therapeutic interventions

Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance

Intermittent hypoxia in obstructive sleep apnoea mediates insulin resistance through adipose tissue inflammation

Role of the inflammasome in insulin resistance and type 2 diabetes mellitus

The Role of Hypothalamic Microglia in the Onset of Insulin Resistance and Type 2 Diabetes: A Neuro-Immune Perspective

Ceramide and ceramide 1-phosphate in health and disease

Adipocyte Ceramides—The Nexus of Inflammation and Metabolic Disease

Inflammation, stress, and diabetes

Inflammation, stress, and diabetes

BACK TO MENU

EPIGENITICS

OPEN TEXT

VIDEO: Diabetes and Insulin: A Triumph for Recombinant DNA Technology

β-Hydroxybutyrate as an epigenetic modifier: Underlying mechanisms and implications

Ketone Bodies as Metabolites and Signalling Molecules at the Crossroad between Inflammation and Epigenetic Control of Cardiometabolic Disorders

BACK TO MENU

AUTOPHAGY

AUTOPHAGY1

Autophagy serves as an indispensable process for cellular homeostasis involved in immunity, inflammation, and metabolism. Either excessive or defective autophagy may be associated with human metabolic diseases, indicating the unique role of autophagy in the regulation of metabolic homeostasis. Diabetes mellitus is featured by hyperglycemia, oxidative stress and mitochondrial injury, all of which are under the tight regulation of autophagy. Autophagy regulates normal function of pancreatic Beta cells and insulin-target tissues, such as liver, skeletal muscle, and adipose tissue. With loss of autophagy homeostasis, damaged mitochondria accumulate, leading to an overt rise in ROS. Mice deficient in autophagy gene in Beta-cells exhibited accumulation of damaged organelles, injured mitochondria, increases in ROS and reduction in glucose-stimulated insulin secretion

Autophagy as an Emerging Target in Cardiorenal Metabolic Disease: from Pathophysiology to Management

Apoptosis and Autophagy: regulatory connections between two supposedly different processes

Apoptosis: A Review of Programmed Cell Death

Apoptosis in pancreatic β-islet cells in Type 2 diabetes

Pancreatic β Cell Mass Death

Physiological and pathological characteristics of vascular endothelial injury in diabetes and the regulatory mechanism of autophag

APOPTOSIS

The ménage à trois of autophagy, lipid droplets and liver disease

Role of Dietary Fructose and Hepatic de novo Lipogenesis in Fatty Liver Disease

Fructose Consumption, Lipogenesis, and Non-Alcoholic Fatty Liver Disease

BACK TO MENU



METHYLGLYOXAL

GLU3

Abstract: Methylglyoxal attenuates insulin signaling and downregulates the enzymes involved in cholesterol biosynthesis:
Methylglyoxal (MG) is a highly reactive dicarbonyl known to be elevated under the hyperglycemic conditions of diabetes and is implicated in the development of diabetic complications.


Methylglyoxal, a highly reactive dicarbonyl compound, as a threat for blood brain barrier integrity

Methylglyoxal and Its Adducts: Induction, Repair, and Association with Disease

Methylglyoxal and Advanced Glycation End products: Insight of the regulatory machinery affecting the myogenic program and of its modulation by natural compounds

Formation of glyoxal, methylglyoxal and 3-deoxyglucosone in the glycation of proteins by glucose

Methylglyoxal and insulin resistance in berberine-treated type 2 diabetic patients

Glycolysis WIKI

Methylglyoxal attenuates insulin signaling and downregulates the enzymes involved in cholesterol biosynthesis

METHYLGLYOXAL, A HIGHLY REACTIVE DICARBONYL COMPOUND, IN DIABETES, ITS VASCULAR COMPLICATIONS, AND OTHER AGE-RELATED DISEASES



BACK TO MENU



OBESITY/LEPTIN/GHRELIN

PI3K88

Obesity is closely associated with various metabolic disorders including dyslipidemia, cardiovascular disease, stroke, insulin resistance, and Type 2 diabetes.
Body weight is controlled by energy intake and energy expenditure. The energy imbalance results in excessive calorie accumulation in the form of triglycerides in adipose tissues, leading to overweight and obesity. In the normal conditions, adipose tissues closely commute with the brain to maintain energy homeostasis and body weight. Adipose tissues secret a variety of humoral factors, collectively called adipokines, to regulate nutrient metabolism. Some adipokines (e.g. leptin) serve as adiposity signals to convey the information about the body energy storage and availability to the brain. The brain, particularly the hypothalamus, senses and integrates these adiposity signals and maintains energy homeostasis and body weight by controlling feeding behavior and energy expenditure.

Obesity Pathogenesis: An Endocrine Society Scientific Statement

Obesity Algorithm

Obesity, diabetes mellitus, and cardiometabolic risk: An Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) 2023

The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis

The PI3K/AKT pathway in obesity and type 2 diabetes.

Metaflammation in glucolipid metabolic disorders: Pathogenesis and treatment

Cellular Senescence in Diabetes Mellitus: Distinct Senotherapeutic Strategies for Adipose Tissue and Pancreatic β Cells

JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age

The PI3K/AKT pathway in obesity and type 2 diabetes

ABSTRACT: Regulation of metabolism by the innate immune system

Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance

POTENTIAL CONTRIBUTORS TO OBESITY

Metabolic Inflammation and Insulin Resistance in Obesity

Impact Of body Mass Index on Cardiopulmonary Outcomes of COVID-19 Hospitalizations Complicated by Severe Sepsis

Characteristics of glucolipid metabolism and complications in novel cluster-based diabetes subgroups:

Fatty Acids, Obesity, and Insulin Resistance: Time for a Reevaluation

Role of adipokines in cardiovascular disease

Obesity and Insulin Resistance: An Ongoing Saga

Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance

The Randle cycle revisited: a new head for an old hat

Randle Cycle

Fructose as a key player in the development of fatty liver disease

High-Fructose Medium-Chain-Trans-Fat Diet Induces Liver Fibrosis & Elevates Plasma Coenzyme Q9 in a Novel Murine Model of Obesity and NASH

The regulation of adipocyte growth in white adipose tissue

Taking aim at belly fat

Adipose Tissue Dysfunction as Determinant of Obesity-Associated Metabolic Complications

Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance

Obesity-Induced Insulin Resistance and Hyperglycemia: Etiological Factors and Molecular Mechanisms

Mechanisms of insulin resistance in obesity

Hyperinsulinemia and Its Pivotal Role in Aging, Obesity, Type 2 Diabetes, Cardiovascular Disease and Cancer

Body Fat Distribution and Insulin Resistance

A Comprehensive Summary of the Current Understanding of the Relationship between Severe Obesity, Metabolic Syndrome, and Inflammatory Status

Fat Cell Numbers in Teen Years Linger for a Lifetime

Obesity, Insulin Resistance, and Type 2 Diabetes: Associations and Therapeutic Implications

Exploring the Pathophysiology of ATP-Dependent Potassium Channels in Insulin Resistance

Muscle and adipose tissue insulin resistance: malady without mechanism

Perspective: A Historical and Scientific Perspective of Sugar and Its Relation with Obesity and Diabetes

Obesity and hypertension: two epidemics or one

Rapid oscillations in omental lipolysis are independent of changing insulin levels in vivo

NAFLD as a continuum: from obesity to metabolic syndrome and diabetes

The role of glucose-6-phosphate dehydrogenase in adipose tissue inflammation in obesity

Abdominal subcutaneous and visceral adipocyte size, lipolysis and inflammation relate to insulin resistance in male obese humans

Single-cell analysis of insulin-regulated fatty acid uptake in adipocytes

Variations in the Size of the Major Omentum Are Primarily Determined by Fat Cell Number

Obesity-induced Changes in Adipose Tissue Microenvironment and Their Impact on Cardiovascular Disease

Fat Cell Size: Measurement Methods, Pathophysiological Origins, and Relationships With Metabolic Dysregulations

Regulation of Lipolysis in Adipocytes

Fructose Consumption, Lipogenesis, and Non-Alcoholic Fatty Liver Disease

Fructose as a key player in the development of fatty liver disease

The Epidemiology of Obesity: A Big Picture

Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance

Targeting PI3K/AKT signaling pathway in obesity

The regulation, function, and role of lipophagy, a form of selective autophagy, in metabolic disorders

The LDL-HDL Profile Determines the Risk of Atherosclerosis: A Mathematical Model

Advanced glycation end products: Key mediator and therapeutic target of cardiovascular complications in diabetes

The Australian Obesity Management Algorithm: A simple tool to guide the management of obesity in primary care

Obesity Society Consensus Statement on Obesity

Obesity Society

A spatiotemporal proteomic map of human adipogenesis


Adipose tissue hypertrophy and insulin resistance

obesity12

Adipose tissue hypertrophy and insulin resistance are closely linked processes that contribute to the development of metabolic disorders, such as obesity and type 2 diabetes. Adipose tissue hypertrophy refers to the increase in size of adipose cells, which can occur in response to an energy imbalance, leading to an accumulation of excess fat.

Insulin resistance, on the other hand, is a condition in which the body’s cells become less responsive to insulin, leading to impaired glucose uptake and increased blood glucose levels. Insulin resistance is a hallmark of metabolic syndrome and is often associated with obesity.

Research suggests that adipose tissue hypertrophy can lead to insulin resistance through several mechanisms:

Adipocyte hypertrophy: As adipose cells increase in size, they become less responsive to insulin, leading to impaired glucose uptake and increased blood glucose levels. Inflammation: Adipose tissue hypertrophy can lead to chronic inflammation, which can impair insulin signaling and increase insulin resistance. Adipokine dysregulation: Adipose tissue produces various hormones, such as leptin and adiponectin, which play a crucial role in glucose and lipid metabolism. Dysregulation of these adipokines can contribute to insulin resistance. Ectopic lipid deposition: Excess fat can accumulate in non-adipose tissues, such as the liver and muscle, leading to insulin resistance.

obesity13

In turn, insulin resistance can exacerbate adipose tissue hypertrophy by:

Reducing glucose uptake: Insulin resistance impairs glucose uptake in adipose tissue, leading to increased glucose levels in the blood and further driving adipose tissue hypertrophy.

Promoting lipolysis: Insulin resistance can stimulate lipolysis, the breakdown of triglycerides into fatty acids and glycerol, leading to increased fat release into the bloodstream and further contributing to adipose tissue hypertrophy.

In summary, adipose tissue hypertrophy and insulin resistance are interrelated processes that can perpetuate each other, leading to the development of metabolic disorders.


obesity14

Antidiabetic and anti-inflammatory effects of branched fatty acid esters of hydroxy fatty acids (FAHFAs). (a) Glucose is transported into adipocytes by the GLUT4 glucose transporter. The increased glucose entry activates the transcription factor ChREBP, thereby enhancing de novo lipogenesis and synthesis of FAHFAs. (b) FAHFAs augment insulin-stimulated glucose transport in adipocytes and glucose-stimulated GLP1 secretion from gut enteroendocrine cells and insulin secretion from pancreatic beta cells. FAHFAs also reduce inflammation by decreasing the production of pro-inflammatory cytokines from macrophages and dendritic cells. ChREBP, carbohydrate response element-binding protein; FAS, fatty acid synthase; GPR120, G protein-coupled receptor 120 [52]. Reproduced with permission from Nature Publishing Group, licence number 3851950681929.

Fat Cell Size: Measurement Methods, Pathophysiological Origins, and Relationships With Metabolic Dysregulations

Adipocyte hypertrophy associates with in vivo postprandial fatty acid metabolism and adipose single-cell transcriptional dynamics

Adipose tissue regulates insulin sensitivity: role of adipogenesis, de novo lipogenesis and novel lipids

Ectopic Fat and Insulin Resistance: Pathophysiology and Effect of Diet and Lifestyle Interventions

Pathophysiology of Obesity

Chronic Adipose Tissue Inflammation Linking Obesity to Insulin Resistance and Type 2 Diabetes

Adipose Tissue Dynamics: Cellular and Lipid Turnover in Health and Disease

Adipose Tissue Distribution, Inflammation and Its Metabolic Consequences, Including Diabetes and Cardiovascular Disease

Obesity, Inflammation, Toll-Like Receptor 4 and Fatty Acids

VIDEO BITTMAN Sarcopenic Obesity

Obese visceral fat tissue inflammation: from protective to detrimental

Reducing sitting time versus adding exercise: differential effects on biomarkers of endothelial dysfunction and metabolic risk

Metabolism Disrupting Chemicals and Metabolic Disorders

Obesity and Severe Obesity Prevalence in Adults: United States, August 2021–August 2023

Obesity, insulin resistance and comorbidities – Mechanisms of association

Trapped fat: Obesity pathogenesis as an intrinsic disorder in metabolic fuel partitioning

Signaling pathways in obesity: mechanisms and therapeutic interventions


Adiponectin signaling and function in insulin target tissues:

OBESITY15

Schematic representation of adiponectin signal transduction pathway implicating a crosstalk with the insulin signaling pathway. Activation of insulin and adiponectin receptors by their respective ligands triggers a cascade of signaling events. Most of the metabolic effects of insulin are mediated by the PI3K/AKT pathway, leading to biological responses that include increased protein synthesis, lipogenesis, glucose uptake and utilization, and glycogen synthesis, and reduced lipolysis and gluconeogenesis. In the case of adiponectin, APPL1 interacts with AdipoR1 or AdipoR2 through its C-terminal PTB and CC domains, and mediates the effects of adiponectin on the activation of multiple pathways including PPAR-α, AMPK, and p38 MAPK. Both AdipoR1 and AdipoR2 are associated with ceramidase activities that are activated upon adiponectin binding. One key binding partner of IRS1/2 is APPL1, which promotes IRS1/2 binding to the insulin receptor and enhances insulin signaling transduction. This crosstalk between insulin and adiponectin signaling pathways is a major mechanism by which adiponectin sensitizes insulin action in insulin target tissues.

Adiponectin shows its effects after binding to specific receptors, AdipoR1 and AdipoR2. Various tissues, including skeletal muscle, liver, and the cardiovascular system show expression of these receptors, which can affect the secretion of adiponectin [51]. Adiponectin receptors are expressed in skeletal muscle. When adiponectin binds to these receptors, it increases insulin sensitivity, which leads to improved glucose uptake by muscle cells and ultimately regulates blood glucose levels [52]. Adiponectin receptors are also expressed in the liver, which suppresses the production of excess glucose. Moreover, adiponectin promotes lipid breakdown in the liver, which prevents excessive fat accumulation and improves insulin sensitivity [53]. Adiponectin exerts anti-inflammatory actions on the cardiovascular system, which results in reducing inflammation in blood vessel walls and preventing atherosclerosis [54].

Role of adipokines in cardiovascular disease

Adiponectin, a Therapeutic Target for Obesity, Diabetes, and Endothelial Dysfunction

Adiponectin: Structure, Physiological Functions, Role in Diseases, and Effects of Nutrition

ADIPONECTIN LEVELS AND THE RISK OF HYPERTENSION

Hypoadiponectinemia as a Predictor for the Development of Hypertension

Role of adipokines in cardiovascular disease

Lipogenesis inhibitors: therapeutic opportunities and challenges

Adiponectin in insulin resistance:

Adiponectin signaling and function in insulin target tissues

Role of adipokines in cardiovascular disease


LEPTIN

Leptin is a protein that is derived primarily from white adipose tissue (WAT) [38]. It suppresses appetite and increases energy expenditure by repressing anabolic neuronal circuits and activating catabolic neuronal circuits. In addition, leptin levels are affected by nutriture [39]. Leptin-mediated appetite and energy homeostasis are associated with the progression of IR [40]. Furthermore, a state called leptin resistance, which was disputed lately by the concept of hypothalamic leptin insufficiency, is often observed in the obese individuals, and weight loss simultaneously reduces serum leptin levels. This suggests that leptin might have a role in regulating IR. Consistent with this, the stimulation of PI3K signaling by leptin is essential for modulating glucose metabolism and the function of pancreatic β-cells [31–42]. It is likely that an increased concentration of leptin, an anti-inflammatory cytokine, during inflammation in AT is associated with leptin resistance in obese individuals. Interestingly, leptin was recommended as a biomarker for in utero insulin resistance based on the link between maternal and fetal leptin and IR [43, 44]. Leptin is a potential treatment for IR because it improves glycometabolism, insulin sensitivity, and lipometabolism [45, 46].



Leptin signaling and leptin resistance

Leptin and Obesity: Role and Clinical Implication

Leptin & Leptin Resistance

Metabolic Inflammation and Insulin Resistance in Obesity

Agrarian diet and diseases of affluence – Do evolutionary novel dietary lectins cause leptin resistance?

Recent Advances in the Knowledge of the Mechanisms of Leptin Physiology and Actions in Neurological and Metabolic Pathologies

Food for Thought: Leptin and Hippocampal Synaptic Function

Interactions between leptin and insulin resistance in patients with prediabetes, with and without NAFLD

The role of leptin in the control of insulin-glucose axis

Obesity Pathogenesis: An Endocrine Society Scientific Statement

GHRELIN

GRHLIN1

Ghrelin regulates glucose hemostasis by inhibiting insulin secretion and regulating gluconeogenesis/glycogenolysis. Ghrelin signaling decreases thermogenesis to regulate energy expenditure. Ghrelin improves the survival prognosis of myocardial infarction by reducing sympathetic nerve activity. Ghrelin prevents muscle atrophy by inducing muscle differentiation and fusion. Ghrelin regulates bone formation and metabolism by modulating proliferation and differentiation of osteoblasts. Ghrelin is also involved in cancer development and metastasis; ghrelin and GHS-R mRNA are highly expressed in metastatic forms of cancers. Ghrelin is now thought to play a significant role in the regulation of lipid storage in white adipose tissue (WAT). Although acute ghrelin exposure also induces Growth Hormone secretion, the net effect of prolonged ghrelin exposure is increased fat mass. Ghrelin has been reported to enhance adipogenesis, augment fat storage enzyme activity, elevate triglyceride content and reduce fat utilisation/lipolysis.

Ghrelin: much more than a hunger hormone

Ghrelin in Diabetes and Metabolic Syndrome

The role of ghrelin in the regulation of glucose homeostasis

Ghrelin's second life: From appetite stimulator to glucose regulator

Ghrelin’s Relationship to Blood Glucose

Ghrelin – Physiological Functions and Regulation

BACK TO MENU



EXERCIZE & Sleep


OPEN TEXT

AMPK and the biochemistry of exercise: Implications for human health and disease

AMPK and Exercise: Glucose Uptake and Insulin Sensitivity

Skeletal muscle AMPK is not activated during 2 h of moderate intensity exercise at ∼65% urn:x-wiley:00223751:media:tjp14224:tjp14224-math-0001 in endurance trained men

The Energy Sensor AMPK: Adaptations to Exercise, Nutritional and Hormonal Signals

Effects of different doses of exercise and diet-induced weight loss on beta-cell function in type 2 diabetes (DOSE-EX):

An overview of muscle glucose uptake during exercise. Sites of regulation

The physiological regulation of glucose flux into muscle in vivo

Moderate to vigorous physical activity early in the day influences weight management, health outcomes

Sedentary Behavior and Health: Update from the 2018 Physical Activity Guidelines Advisory Committee

Accelerometry-Assessed Latent Class Patterns of Physical Activity and Sedentary Behavior With Mortality

Voluntary Exercise Can Ameliorate Insulin Resistance by Reducing iNOS-Mediated S-Nitrosylation of Akt in the Liver in Obese Rats

Meta-analyses of the Association of Sleep Apnea with Insulin Resistance, and the Effects of CPAP on HOMA-IR, Adiponectin, and Visceral Adipose Fat

Adiponectin, a Therapeutic Target for Obesity, Diabetes, and Endothelial Dysfunction

Exercise rescues mitochondrial coupling in aged skeletal muscle: a comparison of different modalities in preventing sarcopenia

Regulation of Skeletal Muscle Glucose Transport and Glucose Metabolism by Exercise Training

Risk of Incident Obstructive Sleep Apnea Among Patients With Type 2 Diabetes

Three hours of intermittent hypoxia increases circulating glucose levels in healthy adults

Exercise-Induced Autophagy Suppresses Sarcopenia Through Akt/mTOR and Akt/FoxO3a Signal Pathways and AMPK-Mediated Mitochondrial Quality Control

Exercise and Regulation of Carbohydrate Metabolism

Exercise Metabolism: Fuels for the Fire

Regulation of Energy Substrate Metabolism in Endurance Exercise

The importance of exercise for glycemic control in type 2 diabetes

Obesity Alters the Muscle Protein Synthetic Response to Nutrition and Exercise

Making Sense of Muscle Protein Synthesis: A Focus on Muscle Growth During Resistance Training

Exerkines: A Comprehensive Term for the Factors Produced in Response to Exercise

Anabolic Resistance of Muscle Protein Turnover Comes in Various Shapes and Sizes

Optimizing Skeletal Muscle Anabolic Response to Resistance Training in Aging

PETER ATTIA Can you maintain muscle during fasting?

Preserve your muscle mass

ABSTRACT Age related muscle anabolic resistance: inevitable or preventable?

Distribution of dietary protein intake in daily meals influences skeletal muscle hypertrophy via the muscle clock

Is It Time to Reconsider the U.S. Recommendations for Dietary Protein and Amino Acid Intake?

VIDEO Protein synthesis animation 20 MIN.

VIDEO DR. WENDI Neuromuscular Junction and muscle contraction

VIDEO DR. WENDI Muscle Metabolism

Role of Dietary Protein and Muscular Fitness on Longevity and Aging

Minimal Intensity Physical Activity (Standing and Walking) of Longer Duration Improves Insulin Action and Plasma Lipids More than Shorter Periods of Moderate to Vigorous Exercise (Cycling) in Sedentary Subjects When Energy Expenditure Is Comparable

Reducing sitting time versus adding exercise: differential effects on biomarkers of endothelial dysfunction and metabolic risk

The association of sleep with metabolic pathways and metabolites: evidence from the Dietary Approaches to Stop Hypertension

BACK TO MENU



MISC

OBESITY1

THIS IS A HOLDING AREA FOR MISC. ITEMS

Glyceroneogenesis Is the Dominant Pathway for Triglyceride Glycerol Synthesis in Vivo in the Rat

Type 2 diabetes mellitus and bone fragility: Special focus on bone imaging

Type 2 Diabetes STAT PERALS

pdate on the impact of type 2 diabetes mellitus on bone metabolism and material properties

Peroxisome Proliferator-Activated Receptors and Caloric Restriction—Common Pathways Affecting Metabolism, Health, and Longevity

Vitamin D and the risk of dementia and Alzheimer disease

Diabetes Mellitus | Type I and Type II Diabetes Mellitus NINJA NERD

Pancreas: Insulin Function NINJA NERD

Digestion & Absorption of Proteins

Standards of Medical Care in Diabetes—2019 Abridged for Primary Care Providers

The clinical importance of visceral adiposity: a critical review of methods for visceral adipose tissue analysis

Resveratrol inhibits hydrogen peroxide-induced apoptosis in endothelial cells via the activation of PI3K/Akt by miR-126

Resveratrol Inhibits Hydrogen Peroxide-Induced Apoptosis in Endothelial Cells via the Activation of PI3K/Akt by miR-126

Oxytocin and Psychological Factors Affecting Type 2 Diabetes Mellitus

CELL Coupled chemical reactions

The integrative biology of type 2 diabetes

Pathophysiology of Type 2 Diabetes Mellitus

Molecular and metabolic mechanisms of insulin resistance and β-cell failure in type 2 diabetes

The new biology of diabetes

m6A mRNA methylation regulates human β-cell biology in physiological states and in type 2 diabetes

A beta cell subset with enhanced insulin secretion and glucose metabolism is reduced in type 2 diabetes

Trajectories of Glycemia, Insulin Sensitivity and Insulin Secretion Preceding the Diagnosis of Type 2 Diabetes: The Whitehall II Study

AMP-activated Protein Kinase and FoxO Transcription Factors in Dietary Restriction–induced Longevity

The Endocrine Pancreas

Physiology, Pancreas

Physiology, Adenosine Triphosphate

The Multifaceted Pyruvate Metabolism: Role of the Mitochondrial Pyruvate Carrier

KREBS1

Amino Acid Metabolism

Loss of insulin signaling in vascular endothelial cells accelerates atherosclerosis in apolipoprotein E null mice

The Hexosamine Biosynthesis Pathway: Regulation and Function

Chronic glucolipotoxic conditions in pancreatic islets impair insulin secretion due to dysregulated calcium dynamics, glucose responsiveness and mitochondrial activity

Pancreas-Brain Crosstalk

Researching glutamate – induced cytotoxicity in different cell lines

TCF7L2 regulates pancreatic β-cell function through PI3K/AKT signal pathway

Exocytosis Proteins: Typical and Atypical Mechanisms of Action in Skeletal Muscle

Imbalance of Angiogenesis in Diabetic Complications: The Mechanisms

Diabetic Neuropathy of the Retina and Inflammation: Perspectives

Quantifying the Contribution of the Liver to Glucose Homeostasis: A Detailed Kinetic Model of Human Hepatic Glucose Metabolism

Patch clamp electrophysiology

Flavonols as a Potential Pharmacological Intervention for Alleviating Cognitive Decline in Diabetes: Evidence from Preclinical Studies

Plant-based diet may feed key gut microbes

Diabetic Gastroparesis

Association of the triglyceride glucose index as a measure of insulin resistance with mortality and cardiovascular disease

Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation

Clinical implications of diabetes in chronic liver disease: Diagnosis, outcomes and management, current and future perspectives

Differentiation of Diabetes by Pathophysiology, Natural History, and Prognosis

Molecular Mechanisms of Hypothalamic Insulin Resistance

Molecular and Cellular Mechanisms of Cardiovascular Disorders in Diabetes

Autonomic dysfunction, diabetes and metabolic syndrome

Delayed heart rate recovery after exercise predicts development of metabolic syndrome: A retrospective cohort study

Influence of Autonomic Nervous System Dysfunction on the Development of Type 2 Diabetes

Ceramide: A Common Pathway for Atherosclerosis

Emerging Roles of Ceramide in Cardiovascular Diseases

Molecular complexities underlying the vascular complications of diabetes mellitus

Diabetes Drugs

The Effect of Magnesium Deficiency on Neurological Disorders: A Narrative Review Article

Hypertension & Anti-hypertensive drugs DM

Differentiating EPA from EPA/DHA in cardiovascular risk reduction

Eicosapentaenoic-to-Arachidonic Acid Ratio Predicts Mortality and Recurrent Vascular Events in Ischemic Stroke Patients

Antioxidant and neuroprotective actions of resveratrol in cerebrovascular diseases

Insulin resistance - Reference pathway

Berberine-induced glucagon-like peptide-1 and its mechanism for controlling type 2 diabetes mellitus

Diabetic neuropathy in the gut: pathogenesis and diagnosis

Treatment of gastrointestinal autonomic neuropathy

Acute caffeine ingestion reduces insulin sensitivity in healthy subjects: a systematic review and meta-analysis

The role of dietary antioxidants in type 2 diabetes and neurodegenerative disorders: An assessment of the benefit profile

The effect of acute caffeine intake on insulin sensitivity and glycemic control in people with diabetes

The physiological effects of caffeine on synaptic transmission and plasticity in the mouse hippocampus selectively depend on adenosine A1 and A2A receptors

Caffeine and the dopaminergic system

Caffeine Alters A2A Adenosine Receptors and Their Function in Human Platelets

Diabetes Drugs

Differential Associations of Oral Glucose Tolerance Test–Derived Measures of Insulin Sensitivity and Pancreatic β-Cell Function With Coronary Artery Calcification and Microalbuminuria in Type 2 Diabetes

Evidence for cephalic phase insulin release in humans: A systematic review and meta-analysis

Use of c-peptide as a measure of cephalic phase insulin release in humans

Ketone bodies mimic the life span extending properties of caloric restriction

Migraine and neuroinflammation: the inflammasome perspective

Risk factors in V-shaped risk associations with all-cause mortality in type 2 diabetes

Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation

The crosstalk between parenchymal cells and macrophages







BACK TO MENU



ALZHEIMERS

ALZHEIMERS

Eating and Swallowing Issues in Persons with Alzheimer’s Disease: Impairment to Treatment

Migraine and neuroinflammation: the inflammasome perspective

Inflammation context in Alzheimer’s disease, a relationship intricate to define

Migraine and neuroinflammation: the inflammasome perspective

Migraine and Alzheimer’s Disease

Risk factors for Alzheimer's disease

BACK TO MENU



POLYOL PATHWAY

POLY2

Increased Polyol (Sorbitol) Pathway Flux The polyol pathway (or sorbitol-aldose reductase pathway) is a two-step process that converts glucose to fructose [57]. In this pathway, glucose is reduced to sorbitol, which is afterward oxidized to fructose. Aldose reductase (AR) is the first enzyme involved. It has a low affinity (high Km) for glucose, and at the normal glucose concentrations found in people without diabetes, metabolism of glucose by this pathway is negligible. But in a hyperglycemic setting (as occurs in uncontrolled diabetes), hexokinase (HK), the rate-limiting enzyme of the common glycolytic pathway (Figure 1), gets saturated and the surplus of glucose enters the polyol pathway, where AR reduces it to sorbitol (Figure 2). This reaction oxidizes NADPH (nicotinamide adenine dinucleotide phosphate) to NADP+. Sorbitol dehydrogenase (SDH) can then oxidize sorbitol to fructose, which produces NADH (nicotinamide adenine dinucleotide) from its oxidized form NAD+ [58]. Hexokinase can restore the molecule to the glycolysis pathway by phosphorylating fructose to form fructose-6-phosphate. However, in uncontrolled diabetes with high blood glucose—more than the glycolysis pathway can cope with—the reaction is altered towards the creation of sorbitol [59].
Although the focus of research on AR has been its involvement in the development of diabetes, many studies have shown that besides reducing glucose, AR efficiently reduces oxidative stress-generated lipid aldehydes with Km in the micromolar range (10–30 μM). In comparison the Km of the enzyme for glucose is in the millimolar range (50–100 mM)

Biochemistry, Polyol Or Sorbitol Pathways

POLYOL PATHWAY

Physiological and Pathological Roles of Aldose Reductase

A New Approach to Control the Enigmatic Activity of Aldose Reductase

Redox imbalance stress in diabetes mellitus: Role of the polyol pathway

The Role of the Pentose Phosphate Pathway in Diabetes and Cancer

The role of glucose-6-phosphate dehydrogenase in adipose tissue inflammation in obesity

BACK TO MENU



Hexosamines and Insulin Resistance

OPEN TEXT

The Hexosamine Biosynthesis Pathway: Regulation and Function

The Nutrient-Sensing Hexosamine Biosynthetic Pathway as the Hub of Cancer Metabolic Rewiring

Fueling the fire: emerging role of the hexosamine biosynthetic pathway in cancer

Metabolic Stress and Cardiovascular Disease in Diabetes Mellitus: The role of protein O-GlcNAc Modification

The Hexosamine Biosynthesis Pathway: Regulation and Function

Hexosamines, insulin resistance and the complications of diabetes: current status.

Dehydrogenase Activity in Red Blood Cells From Hemolytic and Nonhemolytic Variant Subjects

The Hexosamine Biosynthesis Pathway: Regulation and Function

Hexosamines and Insulin Resistance

BACK TO MENU



PENTOSE PHOSPHATE PATHWAY

The PPP is a central component of metabolism in the majority of single- and multicellular organisms. Despite the pathway is central and evolutionary ancient, it possesses a high level of flexibility, which renders it an attractive target for biotechnology and medicine.
In summary:
1. The main biochemical function of the PPP is the biosynthesis of nucleic-acid and amino-acid sugar phosphate precursors.
2. This function of the PPP is bound to the provision of biochemical reducing equivalents in form of NADPH, which renders the PPP an important player in maintaining redox homeostasis.
3. The PPP is highly flexible, dynamic, and is adapting to varying nutrient supply and stress conditions. This coordinates these functions and is required meet cellular metabolic demands in the constantly changing environment.
4. The PPP is important for biotechnology, as its flexibility can be exploited to tune NADPH production, and for medical research, as the PPP activity is altered by bacterial and eukaryotic parasites during the infection process, when stem cells differentiate, when cancer cells maintain redox homeostasis, and in neurons to sustain energy metabolism.
5. Unveiling the complex regulation of the PPP, which despite 80 years of detailed basic and medical research is still not fully understood, appears hence essential for addressing metabolic adaptation and its consequences on cellular and organismic physiology.

QUOTED FROM: The return of metabolism: biochemistry and physiology of the pentose phosphate pathway

The return of metabolism: biochemistry and physiology of the pentose phosphate pathway

VIDEO Metabolism | Pentose Phosphate Pathway 34 MINUTES

The Role of the Pentose Phosphate Pathway in Diabetes and Cancer

Pentose phosphate pathway

Glucose-6-Phosphate Dehydrogenase: Update and Analysis of New Mutations around the World

Inflammation, glucose, and vascular cell damage: the role of the pentose phosphate pathway

Targeting the Pentose Phosphate Pathway in Syndrome X-related Cardiovascular Complications

The Role of the Pentose Phosphate Pathway in Diabetes and Cancer

BACK TO MENU



(Michaelis Constant)

The kinetic parameters for Glucose-6-phosphate isomerase, also known as phosphoglucose isomerase, are as follows: The ( V_{max} ) (maximum rate) is approximately ( 14.0 \pm 1.3 ) U/mg for glucose 6-phosphate1. The ( K_m ) (Michaelis constant) is about ( 0.14 \pm 0.02 ) mM for glucose 6-phosphate1. These values indicate the enzyme’s efficiency and affinity for glucose 6-phosphate during the isomerization process in glycolysis. Keep in mind that these values can vary depending on the assay conditions and the organism from which the enzyme is derived.

The kinetic parameters for Glucose-6-phosphate dehydrogenase (G6PD) are as follows: The ( K_m ) for glucose-6-phosphate (G6P) is approximately ( 38.3 \mu M )1. The ( K_m ) for NADP+ is about ( 6.51 \mu M )1. The ( k_{cat} ) is around ( 689 s^{-1} )1. These parameters indicate the enzyme’s affinity for its substrates and the rate at which it catalyzes the conversion of glucose-6-phosphate to 6-phosphogluconolactone, the first step in the pentose phosphate pathway. This pathway is crucial for producing NADPH, which is essential for protect

Basics of enzyme kinetics graphs

Enzyme kinetics; what happens at the peak of the Gibbs energy graph?

BACK TO MENU



URIC ACID AND OXALATES

Honey as a Potential Natural Antioxidant Medicine: An Insight into Its Molecular Mechanisms of Action

Fructose Intake, Serum Uric Acid, and Cardiometabolic Disorders:

The Urea Cycle Step by Step

Regulation of uric acid metabolism and excretion

Little known facts about oxalates and their poisonous effects

The urea cycle as a source of nitric oxide implicated in the pathogenesis of insulin-dependent diabetes mellitus

Regulation of the urea cycle enzyme genes in nitric oxide synthesis

Oxalates and its effects on our health

Physiological functions and pathogenic potential of uric acid:

OXALATES1

OXALES2

New insights into the mechanism of substrates trafficking in Glyoxylate/Hydroxypyruvate reductases

Mutations in DHDPSL Are ResponsibleFor Primary Hyperoxaluria Type III

Dietary Oxalate Intake and Kidney Outcomes

Urinary oxalate excretion increases with body size and decreaseswith increasing dietary calcium intake among healthy adults

Calcium intake and urinary stone disease

JUDY CHO OXALATES YOUTUBE

JUDY CHO SUPPLEMENTS YOUTUBE

Could Alzheimer’s disease be a maladaptation of an evolutionary survival pathway mediated by intracerebral fructose and uric acid metabolism?

Uric acid and inflammatory markers

Uric acid activates aldose reductase and the polyol pathway for endogenous fructose and fat production causing development of fatty liver in rats

Physiological functions and pathogenic potential of uric acid:

Serum uric acid and disorders of glucose metabolism: the role of glycosuria

Little known facts about oxalates and their poisonous effects

Oxalates and its effects on our health

What is the relationship between serum uric acid level and insulin resistance?: A case-control study

BACK TO MENU


LIVER

The Metabolic Impact of Nonalcoholic Fatty Liver Disease on Cognitive Dysfunction: A Comprehensive Clinical and Pathophysiological Review

BACK TO MENU



DIABETIC NEUROPATHY


Diabetic neuropathy is a neuro-degenerative disorder that encompasses numerous factors that impact peripheral nerves in the context of diabetes mellitus (DM). Diabetic peripheral neuropathy (DPN) is very prevalent and impacts 50% of diabetic patients. DPN is a length-dependent peripheral nerve lesion that primarily causes distal sensory loss, discomfort, and foot ulceration that may lead to amputation. The pathophysiology is yet to be fully understood, but current literature on the pathophysiology of DPN revolves around understanding various signaling cascades involving the polyol, hexosamine, protein-kinase C, AGE, oxidative stress, and poly (ADP ribose) polymerase pathways. The results of research have suggested that hyperglycemia target Schwann cells and in severe cases, demyelination resulting in central and peripheral sensitization is evident in diabetic patients.

Understanding the role of hyperglycemia and the molecular mechanism associated with diabetic neuropathy and possible therapeutic strategies

Diabetic foot ulcers: Classification, risk factors and management

What Are Diabetic Foot Ulcers?

A Narrative Review of Diabetic Macroangiopathy: From Molecular Mechanism to Therapeutic Approaches

Prevalence of Cognitive Impairment and Associated Factors Among Diabetes Mellitus Patients Attending Follow-up Treatment at Fiche General Hospital, North Ethiopia

The Prevalence of Comorbid Depression in Adults With Diabetes

Impact of diabetes on cognitive function and brain structure

BACK TO MENU



J. Methylation Cycle

What is the Methylation Cycle?

NITRIC OXIDE, METHYLATION & HEART HEALTH

71. MTHFR, VIDEO

72. The important role of Methylation Cycle

Methylation Problems Lead to 100s of Terrible Diseases Suzy Cohan

Treating Methylation: Are We Over-supplementing?

Methyl Group Metabolism in Differentiation, Aging, and Cancer

Understanding the Methylation Cycle

Methyl Cycle NutriGenomics

Homocysteine and Dementia: An International Consensus Statement1

Homocysteine: The Rubik 's Cube of Cardiovascular Risk Factors

Homocysteine and Alzheimer 's disease

Glutathione & N-Acetyl L-Cysteine (NAC)

Is an “Epigenetic Diet” for Migraines Justified? The Case of Folate and DNA Methylation

BACK TO MENU



DENENTIA AND DIABETES

The Association of Late-Life Diabetes Status and Hyperglycemia With Incident Mild Cognitive Impairment and Dementia:

BACK TO MENU



LDL

Fructose Intake, Serum Uric Acid, and Cardiometabolic Disorders

Long-Term Levels of LDL-C and Cognitive Function: The CARDIA Study

Brain Function Is Linked to LDL Cholesterol in Older Adults with Cardiovascular Risk

Long-term levels of LDL-C and cognitive function: The CARDIA Study

Inborn coagulation factors are more important cardiovascular risk factors than high LDL-cholesterol in familial hypercholesterolemia

Association between Low-density lipoprotein cholesterol and occipital periventricular hyperintensities

Total cholesterol and all-cause mortality by sex and age: a prospective cohort study among 12.8 million adults

LDL biochemical modifications: a link between atherosclerosis and aging

Conformational changes in the Niemann–Pick type C1 protein NCR1 drive sterol translocation

BACK TO MENU



S. TASTE BUDS

taste8

taste7

Taste buds: cells, signals and synapses

Gustation (Taste) UNIVERSITY OF OREGON

Recent advances in taste transduction and signaling

Taste Receptors and the Transduction of Taste Signals

Pathway

Taste receptors in the gastrointestinal system

Cephalic phase of insulin secretion in response to a meal is unrelated to family history of type 2 diabetes

Cephalic phase insulin release in healthy humans after taste stimulation?

Is the Use of Artificial Sweeteners Beneficial for Patients with Diabetes Mellitus? The Advantages and Disadvantages of Artificial Sweeteners

Sugar-induced cephalic-phase insulin release is mediated by a T1r2+T1r3-independent taste transduction pathway in mice

How does our sense of taste work?

Taste buds: cells, signals and synapses

A systematic review of the biological mediators of fat taste and smell

Gustatory Signaling in the Periphery: Detection, Transmission, and Modulationof Taste Information



Taste Receptors and the Transduction of Taste Signals

taste9

Transduction mechanisms in a generic taste cell. The apical and basolateral surfaces of the cell are separated by tight junctions. The apical surface contains both channels and G-protein-coupled receptors that are activated by chemical stimuli. The basolateral surface contains voltage-gated Na+, K+, and Ca2+ channels, as well as all the machinery for synaptic transmission mediated by serotonin. Also shown are the relevant second messenger systems and intracellular compartments that store Ca2+. The increase in intracellular Ca2+ either by the activation of voltage-gated Ca2+ channels or via the release from intracellular stores causes synaptic vesicles to fuse and release their transmitter onto receptors on primary sensory neurons.



taste10

The transduction of sweet-tasting compounds involves the activation of G-protein-coupled receptors (GPCRs) on the apical surface of taste cells (see Figure 15.13). The particulars of the cascade depend on a number of factors, including the specifics of the stimulus. In the case of sweeteners such as the saccharides, activation of GPCRs depolarizes taste cells by activating adenylate cyclase, which in turn increases the cAMP concentration that will either directly or indirectly close basolateral K+ channels. Synthetic sweeteners, such as saccharine, activate different GPCRs that in turn activate phospholipase C (PLC) to produce IP3 and DAG. An increase in IP3 raises intracellular Ca2+ concentration, leading to transmitter release. An increase in DAG activates PKA, and PKA in turn phosphorylates and closes basolateral K+ channels, further contributing to this effect. Both of these pathways for the perception of sweetness can co-exist in the same taste cell. There are many chemically distinct classes of bitter-tasting compounds (see Figure 15.13). Some of these are alkaloids, like quinine and caffeine; others are l-amino acids, urea, and even salts like MgSO4. Again, not all of these bitter tastants use the same receptor or transduction pathways. Indeed, about forty new receptors for bitter tastants have been recently cloned, and many are found in the same cells. Bitter- tasting organic compounds typically bind to GPCRs that activate gustducin (a G-protein found in taste cells homologous to transducin in photoreceptors), which in turn activates phosphodiesterase, thus lowering the cyclic nucleotide concentration and closing cyclic nucleotide-gated channels on the basolateral membranes of taste cells. Gustducin-knockout mice exhibit impaired responses to bitter compounds, suggesting that this second messenger is involved in the transduction of bitter tastes. Many bitter tastants bind directly to GCPRs that activate PLC and the production of IP3, leading to an increase in intracellular Ca2+. Given the wide variety of chemical structures that evoke a bitter taste, it is not surprising that a variety of transduction mechanisms are employed. The taste of amino acids fall into both the sweet (d-amino acids) and bitter categories (l-isomers), and also uses a variety of transduction mechanisms. An exception is the amino acid l-glutamate (and its sodium salt), which elicits a quite different taste (see above). The effects of l-glutamate on taste cells involves both ionotropic receptors that activate ion channels, and unusual taste-specific metabotropic glutamate receptors (mGluR4) that are less sensitive to glutamate and that close ion channels through a cAMP-dependent pathway. The overall picture that emerges from these admittedly complicated details is that taste cells have a variety of transduction mechanisms. In general, individual taste cells respond to several types of chemical stimuli. Nevertheless, taste cells also exhibit gustatory selectivity. Like olfactory cells, the lower the threshold concentration for detecting a single tastant, the greater the selectivity of the relevant taste cell. Finally, taste receptor mechanisms also adapt to the ongoing presence of a stimulus, although the mechanisms are not understood. If a chemical is left on the tongue for a sufficient time, it ceases to be perceived (consider saliva, for example). Thus, to obtain the full taste of foods, one must either frequently change the types of foods placed in the mouth or wait a sufficient time between helpings, facts that have long been appreciated by restauranteurs and gourMetabolic Syndrome.

Taste receptors in the gastrointestinal system

Gustatory Signaling in the Periphery: Detection, Transmission, and Modulation of Taste Information

BACK TO MENU



ACTION POTENTIAL


ACTIONPOTENTIAL1

NINJA VIDEO Neurology | Resting Membrane, Graded, Action Potentials

VIDEO KHANACADEMY Sodium-potassium pump

Neuron membrane potentials

GRATED POTENTIAL

RESTING POTENTIAL

RESTING POTENTIAL MECHANISM

Neuron graded potential mechanism

VIDEO KHANACADEMY Sodium-potassium pump

Transmitters

The Neuroscience of Pain

VIDEO Chemical Synapses: Neuronal Signal Transmission

VIDEO The Synapse

VIDEO The Action Potential

VIDEO Myelin and saltatory conduction | Action Potentials in Neurons

Physiology, Action Potential

Synaptic Plasticity

Intro to Neuroscience

VIDEO SUSANNAH HANNAFORD Neurotransmitters

synaptic vesicle cycle

BACK TO MENU



E. NITRIC OXIDE

Nitric Oxide and the Intestinal Epithelial Barrier:

QUANTITATIVE ASPECTS OF NITRIC OXIDE PRODUCTION FROM NITRATE AND NITRITE

THR L-ARGIIINE-NITRIC OXIDE PATHWAY

Quantitative aspects of nitric oxide production from nitrate and nitrite

Nitrate- nitrite- nitric oxide pathway in pulmonary arterial hypertension therapeutics

Physiological chemistry of nitric oxide and its metabolites: implications in inflammation

OPTIMAL NITRIC OXIDE PRODUCTION: ARE COMMON DENTAL HYGIENIC PRACTICES PUTTING US AT RISK FOR HEART DISEASE?

Detrimental Effects of Chronic L-Arginine Rich Food on Aging Kidney

Pharmacology of L-Arginine and L-Arginine-Rich Food

Nitric Oxide Synthesis Pathway Arginine Lowers Blood Pressure

part 1 NATHEN BRYAN NO

PART 2 NATHEN BRYAN NO

Antimicrobial effects of a stannous fluoride toothpaste in distinct oral microenvironments

Therapeutic value of stimulating the nitrate-nitrite-nitric oxide pathway to attenuate oxidative stress and restore nitric oxide bioavailability in cardiorenal disease

Nitric oxide: a regulator of mucosal defense and injury

The role of nitric oxide on endothelial function

Elimination of cardiac arrhythmias using oral taurine with L-arginine with case histories: Hypothesis for nitric oxide stabilization of the sinus node

Protein Metabolism

Nathan S. Bryan Ph.D. Co-Founder, Inventor humann Company

Nitric Oxide and Peroxynitrite in Health and Disease

Nitric oxide and arterial disease

Fluoride Depletes Acidogenic Taxa in Oral but Not Gut Microbial Communities in Mice

Nitric Oxide and Apoptosis Induced in Peyer 's Patches by Attenuated Strains of Salmonella enterica Serovar Enteritidis

Characterization of the L-Arginine/Nitric Oxide Pathway and Oxidative Stress in Pediatric Patients with Atopic Diseases>/a>

VIDEO: Nitric Oxide Synthesis Pathway Arginine Lowers Blood Pressure

Ethanol Metabolism and Effects: Nitric Oxide and its Interaction

Ethyl nitrite is produced in the human stomach from dietary nitrate and ethanol, releasing nitric oxide at physiological pH: potential impact on gastric motility

Metabolic Effects of Dietary Nitrate in Health and Disease

VIDEO: Interview with Gold Medal Olympian, Dr. Caldwell B. Esselstyn

Functional Nitric Oxide Nutrition: What are we Missing in our Diets? VIDEO

Arginine: beyond protein

Neuronal Nitric Oxide Synthase

The manifold roles of protein S-nitrosylation in the life of insulin

Regulatory role for the arginine–nitric oxide pathway in metabolism of energy substrates

Systemic Insulin Resistance and Metabolic Perturbations in Chow Fed Inducible Nitric Oxide Synthase Knockout Male Mice: Partial Reversal by Nitrite Supplementation

Regulation of carbohydrate metabolism by nitric oxide and hydrogen sulfide: Implications in diabetes

BACK TO MENU



Nitric Oxide Synthase:

Inducible Nitric Oxide Synthase: Regulation, Structure, and Inhibition

Regulatory role for the arginine-nitric oxide pathway in metabolism of energy substrates

Inducible Nitric Oxide Synthase

Metabolism via arginase or nitric oxide synthase: two competing arginine pathways in macrophages

nducible Nitric-oxide Synthase Is Regulated by the Proteasome Degradation Pathway*

Neuronal Nitric Oxide Synthase in Vascular Physiology and Diseases

Nitric oxide synthases: regulation and function

Inducible nitric oxide synthase: Regulation, structure, and inhibition

Inducible Nitric Oxide Synthase (iNOS) Is a Novel Negative Regulator of Hematopoietic Stem/Progenitor Cell Trafficking

High Glucose Attenuates Protein S-Nitrosylation in Endothelial Cells: Role of Oxidative Stress

S-Nitrosylation: An Emerging Paradigm of Redox Signaling

The manifold roles of protein S-nitrosylation in the life of insulin

Association with Nitric Oxide Synthase on Insulin Secretory Granules Regulates Glucokinase Protein Levels

BACK TO MENU



Action Potential Videos

PLASTICITY

Neuroscience basics: Synaptic transmission - Chemical synapse, Animation

Action Potential in Neurons, Animation.

Neuromuscular Junction, Animation

Overview of the Nervous System, Animation

Neuroscience Basics: Neuroglia Functions, Animation.

Membrane Potential, Equilibrium Potential and Resting Potential, Animation

Membrane Transport, Animation

Long Term Potentiation and Memory Formation, Animation

Cranial Nerves: Basic Anatomy, Functions, Effects of Damage, and Clinical Tests, Animation

Cranial Nerves: Basic Anatomy, Functions, Effects of Damage, and Clinical Tests, Animation

How Synapses Work





BACK TO MENU



Metabolic Diseases are divided into three types—prediabetes, metabolic syndrome (Metabolic Syndrome), and diabetes mellitus (DM)—according to the organ compromise and/or the severeness of the disease by itself. Cellular respiration is how cells get energy from glucose. The process has three main parts:
1. Glycolysis happens in the cytosol and breaks glucose into two pyruvate, producing 2 ATPs and 2 NADHs. 2. The Krebs cycle occurs in the mitochondrial matrix, where pyruvate is turned into acetyl-CoA, which then goes through a series of reactions, producing ATP, NADH, and FADH2.
3. Finally, oxidative phosphorylation uses NADH and FADH2 to create a proton gradient, which helps make more ATP.

The body is a complex organism, and as such, it takes energy to maintain proper functioning. Adenosine triphosphate (ATP) is the source of energy for use and storage at the cellular level.
The majority of ATP synthesis occurs in cellular respiration within the mitochondrial matrix: generating approximately thirty-two ATP molecules per molecule of glucose that is oxidized. ATP is consumed for energy in processes including ion transport, muscle contraction, nerve impulse propagation, substrate phosphorylation, and chemical synthesis. These processes, as well as others, create a high demand for ATP. As a result, cells within the human body depend upon the hydrolysis of 100 to 150 moles of ATP per day to ensure proper functioning.



Breaking down glucose: Cellular respiration As an example of an energy-releasing pathway, let’s see how one of your cells might break down a sugar molecule (say, from that candy you had for dessert). Many cells, including most of the cells in your body, get energy from glucose ( C6H12O6 in a process called cellular respiration. During this process, a glucose molecule is broken down gradually, in many small steps. However, the process has an overall reaction of:
C6H12O6 + O2 →6 CO2 + 6 H2O + ENERGY ATP


Breaking down glucose releases energy, which is captured by the cell in the form of adenosine triphosphate, or ATP. ATP is a small molecule that gives cells a convenient way to briefly store energy. Once it's made, ATP can be used by other reactions in the cell as an energy source. Much as we humans use money because it’s easier than bartering each time we need something, so the cell uses ATP to have a standardized way to transfer energy. Because of this, ATP is sometimes described as the “energy currency” of the cell.

The main source of energy for the body is glucose. Its low blood concentrations can cause seizures, loss of consciousness and death. Long lasting high glucose levels can cause blindness, renal failure, cardiac and peripheral vascular disease, and neuropathy. Blood glucose concentrations need to be maintained within narrow limits. The process of maintaining blood glucose at a steady state is called glucose homeostasis. This is achieved through a balance of the rate of consumption of dietary carbohydrates, utilization of glucose by peripheral tissues, and the loss of glucose through the kidney tubule. The liver and kidney also play a role in glucose homeostasis. This book aims to provide an overview of blood glucose levels in health and diseases.



Type 2 Diabetes | Nucleus Health


Figure 1 Neuroinflammation and migraine. Stimulation of the trigeminal neurons causes the release of neuropeptides, including CGRP, substance P (SP), leading to mast cell degranulation, leukocyte infiltration, glial cell activation, and increased production of inflammatory TNF-α, IL-1, and IL-6 cytokines. Besides, satellite glial cells (SGCs) and trigeminal ganglions (TG) express receptors for CGRP, and CGRP can stimulate intracellular signaling molecules that are relevant to pain, such as cAMP, CREB, MAPK, and ERK. Under the influence of inflammation, activated microglia, T cells, and mast cells can boost the inflammation loop and production of cytotoxic mediators in the CNS


VIDEO

The Australian Obesity Management Algorithm: A simple tool to guide the management of obesity in primary care

VIDEO: The American Diabetes Association’s Standards of Care in Diabetes—2024

COPILOT1

Many Substances Used for Food Processing Are Never Listed on Ingredient Labels