Category: Moms

The role of hormones in energy metabolism

The role of hormones in energy metabolism

Close Diabetic foot care resources Hprmones Filter Hogmones Chemistry This issue AACC Journals Biochemistry Medical Skills Pathology Research Methods in Life Diabetic foot care resources Books Journals Rolf Academic Enter search term Search. Hevener AReichart DJanez AOlefsky J. Osteopontin is an activator of human adipose tissue macrophages and directly affects adipocyte function. Osteocalcin reverses endoplasmic reticulum stress and improves impaired insulin sensitivity secondary to diet-induced obesity through nuclear factor-κB signaling pathway. Li, X. Reue KDwyer JR. The role of hormones in energy metabolism

Video

Glycogen metabolism

The role of hormones in energy metabolism -

RANKL regulates metabolism by binding with RANK on the cell membrane. In addition, MAT can also secrete the adipokines leptin and adiponectin.

Leptin and adiponectin combine with ADIPOR and OBR on the cell membrane of peripheral tissue, regulating cellular sugar metabolism.

They also act on the central nervous system, leading to reduced food intake and energy consumption. Leptin, which is encoded by the obesity gene Ob and secreted primarily by WAT, is directly related to obesity.

Mutations of the leptin gene lead to obesity. At present, the regulatory mechanism of leptin on energy metabolism has been clearly studied. It was recently discovered that MAT also expresses leptin.

Additionally, leptin expressed in bone MAT can directly act on leptin receptors in bone and affect bone growth by activating FGF23 and regulating the secretion of osteocalcin. There is a complex relationship between bone and energy metabolism.

The main action of circulating endocrine factors is not limited to regulating energy metabolism but affects bone metabolism and remodeling. Insulin is an important hormone regulating energy metabolism in the human body, and its receptor exists in almost all cells.

Insulin promotes glycogen, fat, and protein synthesis by binding to IRs and ultimately lowers blood sugar. demonstrated in mice that when osteoblasts lack IR, osteoporosis can easily develop after birth. However, it may also lead to bone fragility by increasing cortical osteoporosis or other bone structural defects.

The regulation of bone by insulin can affect whole-body energy metabolism. Insulin can promote osteoblast differentiation and glucose uptake by increasing the expression of Glut4 on the cell membrane.

The absence of this pathway will lead to peripheral insulin resistance, which is mainly manifested by an increase in systemic insulin levels and a decrease in insulin sensitivity.

However, no significant phenotypes were found in skeletal muscle cells and adipocytes. Therefore, the decrease in insulin absorption by bone may lead to systemic insulin resistance, which proves the influence of insulin and bone on whole-body energy metabolism from another aspect.

Vitamin D is a steroid hormone that plays a vital role in maintaining bone metabolism and calcium homeostasis. On the other hand, when the serum calcium level declines, VDR in osteoblasts can enhance bone resorption, thereby mobilizing bone calcium into the blood to maintain serum calcium homeostasis.

The relationship between vitamin D and metabolic diseases such as obesity, diabetes, and NAFLD is a trend in research. Adipocytes are the main storage sites of vitamin D and express active vitamin D and VDR.

Vitamin D supplementation can reduce the risk of elevated blood glucose and insulin resistance. Parathyroid hormone PTH can directly affect bone and kidney to regulate calcium and phosphorus metabolism.

One of the key mechanisms of PTH in regulating calcium homeostasis is stimulating bone remodeling. It not only promotes bone resorption and mobilizes bone calcium into blood but also promotes osteogenesis and ameliorates osteoporosis. PTH1R is expressed on the surface of osteoblasts and many bone cells.

In addition to regulating systemic calcium and phosphorus metabolism, PTH also has some effects on energy metabolism, which may be related to the level of vitamin D. It was found that the level of blood sugar increased significantly after parathyroidectomy and decreased after PTH administration, while the secretion of insulin remained unchanged.

also demonstrated that PTH could reduce the blood sugar level in obese type 2 diabetic rats without changing the serum insulin level. One piece of evidence is that the insulin sensitivity index is negatively correlated with plasma PTH levels. PTH induces lipolysis of adipocytes by activating the cAMP-PKA pathway, leading to increased serum cholesterol and triglyceride concentrations in mice.

In addition to reproductive function, estrogen also regulates bone metabolism and energy metabolism. Lack of estrogen increases the risk of metabolic syndromes such as obesity and type 2 diabetes mellitus. In the central nervous system, activation of ERs in the ventromedial hypothalamus and arcuate nucleus can control dietary intake.

Postmenopausal estrogen deficiency can lead to osteoporosis, which is due to estrogen inducing osteoclast apoptosis through ERs and protecting bone through the inflammatory mediators RANKL and Scl. It has been known for a long time that skeletal health is closely related to the overall metabolism of the individual.

For example, people with diabetes have a greater risk of developing osteoporosis than normal individuals, , and a low body mass index causes bone loss. Later, through the discovery of osteocalcin, the first bone-derived factor that regulates energy metabolism, researchers found that bone could also regulate energy metabolism throughout the body.

Some findings showed that bone-derived factors, such as Lcn2 secreted by osteoblasts and adiponectin and leptin secreted by MAT, could cross the blood—brain barrier and act on the central nervous system, which is one of the discoveries in recent years.

Further study on bone-derived factors and their interactions will help to understand the fundamentals of the central nervous system for energy balance. Bone-derived exosomes are also a new research area in recent years and have been found to regulate bone itself and other tissues.

The effect of bone-derived factors on the regulation of energy metabolism-related organs is complicated. This may be due to the different distributions and proportions of receptors on the surfaces of other cells so that these factors have more than one regulatory pathway.

Moreover, sometimes the results of experiments on animals and cell cultures in vitro may be different from those obtained from human subjects, indicating that the human body has a more complex internal environment, which impacts the results.

Bone-derived factors may be affected by a variety of factors in the processes involved in secretion, circulation and function, including genetic background diversity, age and health status of the human body. For example, decarboxylation of OCN is vitamin K dependent, so the vitamin K concentration in the human body will affect the physiological function of osteocalcin.

One of the key points in studying the regulation of bone-derived factors on energy metabolism is to confirm whether the change in the levels of those factors is the cause but not the result of altered energy metabolism.

Because of the close relationship between bone metabolism and whole-body energy metabolism, the levels of factors secreted by bone are likely to be determinants of energy homeostasis and metabolism.

Second, it is of interest to determine whether those factors that play a crucial role in other tissues and organs are from bone. This could be easily determined for some factors, such as osteocalcin and FGF23, which are mostly secreted by bone cells. For other factors expressed in multiple tissues and organs, most of them need to be assessed through genetic engineering techniques by using animal models Table 2.

However, at present, not all bone-derived factors may possess the necessary gene-specific knockout animal models applicable to energy metabolism regulation experiments, such as BMP and OPN. Another reason is that although some autocrine factors in bone cannot be secreted into the blood circulation to act on other organs, they can indirectly affect systemic metabolism by regulating bone metabolism, which should not be ignored.

In view of the complex regulation of whole-body energy metabolism by bone, the clinical application of bone-derived factors will become one of the future research directions. For example, studying the role of bone-derived factors in aging, which is a severe problem facing society, may explain some diseases.

In the process of human aging, bone metabolism and whole-body metabolism are continually changing. Aging may lead to metabolic disorders and affect bone metabolism.

Macroscopically, the aging of bone is characterized by osteoporosis and a decrease in organic and inorganic components. At the same time, its significance is marked by a decrease in bone cells and the transformation of BMSCs into MAT and osteoblasts. Bone is an important motor organ, so exercise can have a significant impact on bone metabolism.

Therefore, exercise therapy may be used to treat or prevent metabolic diseases, especially those related to aging, in the future Fig. The network relationship between bone and whole-body energy metabolism.

Aging is one of the main causes of bone and whole-body metabolism disorder, and exercise training can alleviate it. Changes in bone metabolism will affect multiple organs and tissues, including the brain a potential research direction in the future , and lead to metabolic diseases.

Metabolic diseases, as well as changes in circulating endocrine factors, can in turn affect bone. On the other hand, bone-derived factors can also be used as autocrine factors to regulate their own metabolic state.

The next question is whether we can use bone-derived factors as a marker for the diagnosis or as a new target for the treatment of metabolic diseases and their related complications.

This conjecture has been verified by some research. For example, OPG has been found to be a potential marker for the diagnosis of diabetes in postmenopausal women, and experiments have explored whether regulating the proportion of OPG-RANKL-RANK signaling can interfere with the process of cardiovascular complications noted in patients with diabetes.

Sommerfeldt, D. Biology of bone and how it orchestrates the form and function of the skeleton. Spine J. Article PubMed Central PubMed Google Scholar. Bronner, F.

Extracellular and intracellular regulation of calcium homeostasis. World J. Article CAS Google Scholar. Askmyr, M. Effects of the bone marrow microenvironment on hematopoietic malignancy. Bone 48 , — Article PubMed Google Scholar. Abarrategi, A. et al. Modeling the human bone marrow niche in mice: from host bone marrow engraftment to bioengineering approaches.

Article CAS PubMed Central PubMed Google Scholar. Kajimura, D. Adiponectin regulates bone mass via opposite central and peripheral mechanisms through FoxO1. Cell Metab.

Karsenty, G. Convergence between bone and energy homeostases: leptin regulation of bone mass. Article CAS PubMed Google Scholar. Lecka-Czernik, B. Diabetes, bone and glucose-lowering agents: basic biology. Diabetologia 60 , — Andrukhova, O.

Fgf23 and parathyroid hormone signaling interact in kidney and bone. Cell Endocrinol. Cai, X. DOK3 modulates bone remodeling by negatively regulating osteoclastogenesis and positively regulating osteoblastogenesis.

Bone Min. Kalbasi Anaraki, P. Matsuoka, K. Osteoclast-derived complement component 3a stimulates osteoblast differentiation. Shimada, T. FGF is a potent regulator of vitamin D metabolism and phosphate homeostasis.

Gupte, A. Osteocalcin protects against nonalcoholic steatohepatitis in a mouse model of metabolic syndrome. Endocrinology , — Article PubMed Central CAS PubMed Google Scholar. Du, J. Osteocalcin improves nonalcoholic fatty liver disease in mice through activation of Nrf2 and inhibition of JNK.

Endocrine 53 , — Mizokami, A. Osteocalcin induces release of glucagon-like peptide-1 and thereby stimulates insulin secretion in mice. PLoS ONE 8 , e Otani, T. Signaling pathway for adiponectin expression in adipocytes by osteocalcin.

Cell Signal 27 , — Booth, S. The role of osteocalcin in human glucose metabolism: marker or mediator? Hauschka, P. Osteocalcin and matrix Gla protein: vitamin K-dependent proteins in bone.

Ducy, P. Increased bone formation in osteocalcin-deficient mice. Nature , — Ishida, M. Osteocalcin fragment in bone matrix enhances osteoclast maturation at a late stage of osteoclast differentiation.

Nikel, O. Structural role of osteocalcin and osteopontin in energy dissipation in bone. Hu, C. High glucose triggers nucleotide imbalance through O-GlcNAcylation of key enzymes and induces KRAS mutation in pancreatic cells.

e10 Lee, N. Endocrine regulation of energy metabolism by the skeleton. Cell , — Malashkevich, V. X-ray crystal structure of bovine 3 Glu-osteocalcin.

Biochemistry 52 , — Rached, M. FoxO1 expression in osteoblasts regulates glucose homeostasis through regulation of osteocalcin in mice. Guedes, J. Osteocalcin improves insulin resistance and inflammation in obese mice: Participation of white adipose tissue and bone.

Bone , 68—82 Levinger, I. The effects of muscle contraction and recombinant osteocalcin on insulin sensitivity ex vivo. Lin, X.

Recombinant uncarboxylated osteocalcin per se enhances mouse skeletal muscle glucose uptake in both extensor digitorum longus and soleus muscles. Front Endocrinol. Article Google Scholar. Pi, M. Evidence for osteocalcin binding and activation of GPRC6A in β-cells. Sanchez-Gurmaches, J. Brown Fat AKT2 is a cold-induced kinase that stimulates ChREBP-mediated de novo lipogenesis to optimize fuel storage and thermogenesis.

e6 Gao, J. Peptides 86 , 72—79 Drucker, D. Mechanisms of action and therapeutic application of glucagon-like peptide Oral administration of osteocalcin improves glucose utilization by stimulating glucagon-like peptide-1 secretion. Bone 69 , 68—79 Zhou, B. Osteocalcin reverses endoplasmic reticulum stress and improves impaired insulin sensitivity secondary to diet-induced obesity through nuclear factor-κB signaling pathway.

Guo, Q. Undercarboxylated osteocalcin reverts insulin resistance induced by endoplasmic reticulum stress in human umbilical vein endothelial cells. Jung, C. Metabolism 62 , — Kalucka, J. Quiescent endothelial cells upregulate fatty acid β-oxidation for vasculoprotection via redox homeostasis.

e13 Li, X. Hallmarks of endothelial cell metabolism in health and disease. Hill, H. Carboxylated and uncarboxylated forms of osteocalcin directly modulate the glucose transport system and inflammation in adipocytes. Clemens, T. The osteoblast: an insulin target cell controlling glucose homeostasis.

De Toni, L. Osteocalcin, a bone-derived hormone with important andrological implications. Andrology 5 , — Cell Death Dis. Li, Q. Cell Biol. Mottillo, E. SERCA2b cycles its way to UCP1-independent thermogenesis in beige fat.

Deppermann, C. Macrophage galactose lectin is critical for Kupffer cells to clear aged platelets. Wellendorph, P. Molecular cloning, expression, and sequence analysis of GPRC6A, a novel family C G-protein-coupled receptor.

Gene , 37—46 Ackerman, S. GPRC6A mediates responses to osteocalcin in β-cells in vitro and pancreas in vivo. Fu, A. Role of AMPK in pancreatic beta cell function. Ardestani, A. mTORC1 Signaling: A Double-Edged Sword in Diabetic β Cells. Karmaus, P. Critical roles of mTORC1 signaling and metabolic reprogramming for M-CSF-mediated myelopoiesis.

Lin, S. AMPK: sensing glucose as well as cellular energy status. GPRC6A: Jack of all metabolism or master of none. Diegel, C. An osteocalcin-deficient mouse strain without endocrine abnormalities.

PLoS Genet. Choi, H. Vitamin K2 supplementation improves insulin sensitivity via osteocalcin metabolism: a placebo-controlled trial. Diabetes Care 34 , e Pollock, N.

Lower uncarboxylated osteocalcin concentrations in children with prediabetes is associated with beta-cell function. Choudhury, A. Role of adiponectin in mediating the association of osteocalcin with insulin resistance and type 2 diabetes: a cross sectional study in pre- and post-menopausal women.

Martin, T. Simonet, W. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell 89 , — Boyce, B. Biology of RANK, RANKL, and osteoprotegerin.

Arthritis Res Ther. Nakashima, T. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Chen, C. MiR regulates osteoclastogenesis via targeting RANK. Kondegowda, N. Osteoprotegerin and denosumab stimulate human beta cell proliferation through inhibition of the receptor activator of NF-κB ligand pathway.

Sacco, F. Phosphoproteomics reveals the GSK3-PDX1 axis as a key pathogenic signaling node in diabetic islets. e3 Musialik, K. The relation between osteoprotegerin, inflammatory processes, and atherosclerosis in patients with metabolic syndrome.

Med Pharm. CAS Google Scholar. Monseu, M. Osteoprotegerin levels are associated with liver fat and liver markers in dysmetabolic adults. Diabetes Metab. Cappel, D. Pyruvate-carboxylase-mediated anaplerosis promotes antioxidant capacity by sustaining TCA cycle and redox metabolism in liver.

e8 Bilgir, O. Minerva Endocrinol. Suliburska, J. The association of insulin resistance with serum osteoprotegerin in obese adolescents. Biochem 69 , — Stekovic, S. Alternate day fasting improves physiological and molecular markers of aging in healthy, non-obese humans.

Ou, D. TNF-related apoptosis-inducing ligand death pathway-mediated human beta-cell destruction. Diabetologia 45 , — Chamoux, E. Osteoprotegerin decreases human osteoclast apoptosis by inhibiting the TRAIL pathway.

Cell Physiol. Vaccarezza, M. Knudsen, J. β cell dysfunction in type 2 diabetes: drained of energy? Schrader, J. Cytokine-induced osteoprotegerin expression protects pancreatic beta cells through p38 mitogen-activated protein kinase signalling against cell death.

Diabetologia 50 , — Taylor, R. Remission of human type 2 diabetes requires decrease in liver and pancreas fat content but is dependent upon capacity for β cell recovery. Lacombe, J. In vivo analysis of the contribution of bone resorption to the control of glucose metabolism in mice.

Niu, Y. Plasma osteoprotegerin levels are inversely associated with nonalcoholic fatty liver disease in patients with type 2 diabetes: a case-control study in China. Metabolism 65 , — Samuel, V. Nonalcoholic fatty liver disease as a nexus of metabolic and hepatic diseases.

Ayaz, T. The relation between carotid intima media thickness and serum osteoprotegerin levels in nonalcoholic fatty liver disease. Expert Rev. Cardiovasc Ther. Kiechl, S. Blockade of receptor activator of nuclear factor-κB RANKL signaling improves hepatic insulin resistance and prevents development of diabetes mellitus.

Lacey, D. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 93 , — Fan, Y. Parathyroid hormone directs bone marrow mesenchymal cell fate. Franzén, A. Isolation and characterization of two sialoproteins present only in bone calcified matrix.

Gimba, E. Human osteopontin splicing isoforms: known roles, potential clinical applications and activated signaling pathways. Cancer Lett. Luukkonen, J. Osteoclasts secrete osteopontin into resorption lacunae during bone resorption. Oldberg, A. Cloning and sequence analysis of rat bone sialoprotein osteopontin cDNA reveals an Arg-Gly-Asp cell-binding sequence.

Natl Acad. USA 83 , — Article CAS PubMed PubMed Central Google Scholar. Wang, K. Osteopontin: role in immune regulation and stress responses. Cytokine Growth Factor Rev.

Ge, Q. Osteopontin regulates macrophage activation and osteoclast formation in hypertensive patients with vascular calcification.

Ishijima, M. Enhancement of osteoclastic bone resorption and suppression of osteoblastic bone formation in response to reduced mechanical stress do not occur in the absence of osteopontin.

Chen, Q. An osteopontin-integrin interaction plays a critical role in directing adipogenesis and osteogenesis by mesenchymal. Stem Cells 32 , — Chapman, J. Osteopontin is required for the early onset of high fat diet-induced insulin resistance in mice.

PLoS ONE 5 , e Inoue, H. Role of STAT-3 in regulation of hepatic gluconeogenic genes and carbohydrate metabolism in vivo. Kiefer, F. Neutralization of osteopontin inhibits obesity-induced inflammation and insulin resistance. Diabetes 59 , — Kon, S. Syndecan-4 protects against osteopontin-mediated acute hepatic injury by masking functional domains of osteopontin.

Nuñez-Garcia, M. Osteopontin regulates the cross-talk between phosphatidylcholine and cholesterol metabolism in mouse liver. Lipid Res. Zeyda, M. Osteopontin is an activator of human adipose tissue macrophages and directly affects adipocyte function.

Arafat, H. Osteopontin protects the islets and beta-cells from interleukin-1 beta-mediated cytotoxicity through negative feedback regulation of nitric oxide. Ma, D. A new transkingdom dimension to NO signaling. Wendt, A. PLoS ONE 12 , e Ahmad, R.

Interaction of osteopontin with IL in obese individuals: implications for insulin resistance. Barchetta, I. Increased circulating osteopontin levels in adult patients with type 1 diabetes mellitus and association with dysmetabolic profile.

Carbone, F. Serum levels of osteopontin predict diabetes remission after bariatric surgery. Osteopontin expression in human and murine obesity: extensive local up-regulation in adipose tissue but minimal systemic alterations.

Talat, M. The role of osteopontin in the pathogenesis and complications of type 1 diabetes mellitus in children. Lee, H. Beta cell dedifferentiation induced by IRE1α deletion prevents type 1 diabetes. e5 Marciano, R. Association of alleles at polymorphic sites in the Osteopontin encoding gene in young type 1 diabetic patients.

Warshauer, J. New frontiers in the treatment of type 1 diabetes. You, J. Serum osteopontin concentration is decreased by exercise-induced fat loss but is not correlated with body fat percentage in obese humans.

Israel, D. Heterodimeric bone morphogenetic proteins show enhanced activity in vitro and in vivo. Growth Factors 13 , — Urist, M. Bone: formation by autoinduction. Science , — Celeste, A. Identification of transforming growth factor beta family members present in bone-inductive protein purified from bovine bone.

USA 87 , — Brazil, D. BMP signalling: agony and antagony in the family. Trends Cell Biol. Chen, D. Bone morphogenetic proteins. Growth Factors 22 , — Nohno, T.

Identification of a human type II receptor for bone morphogenetic protein-4 that forms differential heteromeric complexes with bone morphogenetic protein type I receptors.

ten Dijke, P. Ebara, S. Mechanism for the action of bone morphogenetic proteins and regulation of their activity. Spine 27 , S10—S15 Heldin, C.

TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Wang, P. Combined inhibition of DYRK1A, SMAD, and trithorax pathways synergizes to induce robust replication in adult human beta cells. Lu, Q. Tissue Int. Guo, X. Cell Res. Lowery, J. The BMP pathway and its inhibitors in the skeleton.

Yang, M. Maeda, Y. Cell Biochem. Peng, Y. Elsen, M. BMP4 and BMP7 induce the white-to-brown transition of primary human adipose stem cells. Fabbiano, S. Caloric restriction leads to browning of white adipose tissue through type 2 immune signaling. Gustafson, B. BMP4 and BMP antagonists regulate human white and beige adipogenesis.

Diabetes 64 , — Hata, K. Differential roles of Smad1 and p38 kinase in regulation of peroxisome proliferator-activating receptor gamma during bone morphogenetic protein 2-induced adipogenesis. Cell 14 , — Hino, J. Overexpression of bone morphogenetic protein-3b BMP-3b in adipose tissues protects against high-fat diet-induced obesity.

Int J. Hoffmann, J. BMP4 gene therapy in mature mice reduces BAT activation but protects from obesity by browning subcutaneous adipose tissue. Cell Rep. Huang, H. USA , — Kim, S. BMP-9 enhances fibroblast growth factor 21 expression and suppresses obesity.

Acta , — Tseng, Y. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure.

Wang, E. Growth Factors 9 , 57—71 Whittle, A. BMP8B increases brown adipose tissue thermogenesis through both central and peripheral actions. Wang, Q. Reversible de-differentiation of mature white adipocytes into preadipocyte-like precursors during lactation.

Li, C. MicroRNA regulates age-related switch between osteoblast and adipocyte differentiation. Article PubMed PubMed Central Google Scholar. Zhang, R. The role of microRNAs in adipocyte differentiation. Chattopadhyay, T. Biofactors 43 , — Luo, Y. Decreased circulating BMP-9 levels in patients with Type 2 diabetes is a signature of insulin resistance.

Schreiber, I. BMPs as new insulin sensitizers: enhanced glucose uptake in mature 3T3-L1 adipocytes via PPARγ and GLUT4 upregulation. Role of bone morphogenetic protein-9 in the regulation of glucose and lipid metabolism.

FASEB J. Wang, X. New association of bone morphogenetic protein 4 concentrations with fat distribution in obesity and Exenatide intervention on it. Lipids Health Dis. Hodgson, J. Characterization of GDF2 mutations and levels of BMP9 and BMP10 in pulmonary arterial hypertension.

Care Med. Vukicevic, S. Developing human lung and kidney are major sites for synthesis of bone morphogenetic protein-3 osteogenin. Kokubu, N. Yamashita, K. BMP3 expression in the adult rat CNS. Brain Res. Desroches-Castan, A.

Cells 8 , Article CAS PubMed Central Google Scholar. Beenken, A. The FGF family: biology, pathophysiology and therapy.

Drug Discov. Ornitz, D. Fibroblast growth factor signaling in skeletal development and disease. Genes Dev. Emerging structure-function paradigm of endocrine FGFs in metabolic diseases. Trends Pharm. Peng, M. Developments in the study of gastrointestinal microbiome disorders affected by FGF19 in the occurrence and development of colorectal neoplasms.

Geller, S. Tanycytes regulate lipid homeostasis by sensing free fatty acids and signaling to key hypothalamic neuronal populations via FGF21 secretion.

e7 Nishimura, T. Identification of a novel FGF, FGF, preferentially expressed in the liver. Erben, R. Pleiotropic actions of FGF Clinkenbeard, E. Conditional deletion of murine Fgf interruption of the normal skeletal responses to phosphate challenge and rescue of genetic hypophosphatemia.

Bone Miner. Hu, M. Klotho: a novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule. Strewler, G. Angelin, B. Circulating fibroblast growth factors as metabolic regulators-a critical appraisal. Komaba, H.

Klotho expression in osteocytes regulates bone metabolism and controls bone formation. Kidney Int. Rhee, Y. Parathyroid hormone receptor signaling in osteocytes increases the expression of fibroblast growth factor in vitro and in vivo.

Bone 49 , — Kaludjerovic, J. Klotho expression in long bones regulates FGF23 production during renal failure. Kurosu, H. Suppression of aging in mice by the hormone Klotho.

Hesse, M. Ablation of vitamin D signaling rescues bone, mineral, and glucose homeostasis in Fgf deficient mice. Matrix Biol. López, I. Direct and indirect effects of parathyroid hormone on circulating levels of fibroblast growth factor 23 in vivo.

Singh, S. Fibroblast growth factor 23 directly targets hepatocytes to promote inflammation in chronic kidney disease.

Ito, N. Regulation of FGF23 expression in IDG-SW3 osteocytes and human bone by pro-inflammatory stimuli. Mirza, M. Circulating fibroblast growth factor is associated with fat mass and dyslipidemia in two independent cohorts of elderly individuals.

Arterioscler Thromb. Aljohani, A. Hepatic stearoyl CoA desaturase 1 deficiency increases glucose uptake in adipose tissue partially through the PGC-1α-FGF21 axis in mice. Chen, Y. Acute changes of bile acids and FGF19 after sleeve gastrectomy and Roux-en-Y gastric bypass. Lan, T. Mosialou, I.

MC4R-dependent suppression of appetite by bone-derived lipocalin 2. Zhang, J. The role of lipocalin 2 in the regulation of inflammation in adipocytes and macrophages. Flower, D.

Beyond the superfamily: the lipocalin receptors. Biochim Biophys. Jha, M. Diverse functional roles of lipocalin-2 in the central nervous system. Biobehav Rev. Adriaenssens, A. Glucose-dependent insulinotropic polypeptide receptor-expressing cells in the hypothalamus regulate food intake.

Liu, H. Transgenic mice expressing green fluorescent protein under the control of the melanocortin-4 receptor promoter. Guo, H. Lipocalin 2, a regulator of retinoid homeostasis and retinoid-mediated thermogenic activation in adipose tissue.

Lipocalin-2 deficiency impairs thermogenesis and potentiates diet-induced insulin resistance in mice. Yu, B. It also promotes the breakdown of fats in adipose tissue to release fatty acids for energy production.

Cortisol is a steroid hormone produced by the adrenal glands in response to stress. It plays a crucial role in the metabolism of carbohydrates, proteins, and fats. Cortisol promotes the breakdown of proteins in muscle cells to release amino acids for gluconeogenesis, the production of glucose from non-carbohydrate sources.

It also promotes the breakdown of fats in adipose tissue and the release of fatty acids for energy production. Thyroid hormones, produced by the thyroid gland, play a critical role in the regulation of metabolism. They increase the metabolic rate by promoting the production of ATP, the energy currency of cells.

Thyroid hormones also increase the activity of enzymes involved in carbohydrate, protein, and fat metabolism. Leptin is another hormone that plays a crucial role in the regulation of metabolism. It is produced by adipose tissue and is involved in the regulation of energy balance.

Leptin suppresses appetite and increases energy expenditure, promoting weight loss. Ghrelin is a hormone produced by the stomach that stimulates appetite and promotes food intake. It also plays a role in the regulation of energy balance by promoting the release of growth hormone, which increases the breakdown of fats for energy production.

Adiponectin is a hormone produced by adipose tissue that regulates glucose and lipid metabolism. Adiponectin increases insulin sensitivity, promoting glucose uptake by cells for energy production.

It also promotes the breakdown of fats in adipose tissue and the utilization of fatty acids for energy production. In conclusion, hormones play a crucial role in the control of metabolism.

Insulin and glucagon regulate glucose levels in the blood, while cortisol promotes the breakdown of proteins and fats for energy production.

Weight management is a common Polyphenols and bone health for women and men of all ages. Natural fat blocker rkle hormones that also play a critical role Diabetic foot care resources roe weight management enwrgy cortisol, insulin, progesterone, and thyroid hormone triiodothyronine T3. Hormoens an imbalance occurs, it can Te related to metabolidm, Natural fat blocker, illness or stress. Hormones produced by the ovaries, such as estrogen and progesterone, may regulate where fat sits on the body and help determine whether your metabolic engine will use sugar or fat. When a hormone imbalance occurs, women may find they develop extra fat around the middle. Exercise is an essential component to controlling weight, is vital for health, and may reduce some of the problems associated with hormone imbalances. You cannot change fat placement, but you can increase activity to alter your gut flora, change how you extract calories from food and reduce fat storage. University of Missouri-Columbia, USA. You Natural energy sources also merabolism for this editor rolee PubMed Google Scholar. Part of the book series: Advances in Experimental Medicine and Biology AEMB, volume This is a preview of subscription content, log in via an institution to check for access. David M. Klachko, Ralph R. Anderson, Murray Heimberg.

Author: Goltisida

1 thoughts on “The role of hormones in energy metabolism

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com