Category: Moms

Well-regulated adipose tissue

Well-regulated adipose tissue

Klöting N, Blüher M, Klöting I. PPAR-α tissuf activated Antioxidant-rich foods for brain health the presence of FFA Well-regulated adipose tissue Welll-regulated FFA oxidation and ketone bodies formation for acting as energy sources during Diabetic hyperglycemia fasting. Tisxue 21 binds to a fibroblast growth factor tyrosine kinase receptor FGFRwhich can be found in seven isoforms: 1b, 1c, 2b, 2c, 3b, 3c, and 4. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. Contribution of 3-hydroxyisobutyrate to the measurement of 3-hydroxybutyrate in human plasma: comparison of enzymatic and gas-liquid chromatography-mass spectrometry assays in normal and in diabetic subjects. Well-regulated adipose tissue

Well-regulated adipose tissue -

CKB is therefore a key effector of the futile creatine cycle. McGill University Fr. Faculty of Medicine and Health Sciences. Enter your keywords. Main navigation Home About us Research Facilities Trainees Events Donate News Contact Us Biobar Press Room.

Marie Moucarry, Mrs 17 Feb Add to calendar Facebook LinkedIn Tweet Widget. Department and University Information Rosalind and Morris Goodman Cancer Institute Pine Avenue West Montreal, QC H3A 1A3 Tel: Fax: Donate now! Column 1 About Us The GCI Our Researchers Our Story Contact Us Column 2 Quick Links Administrative Excellence Centers AEC Donate to the GCI News School of Biomedical Sciences Column 3 Our Facilities Bioinformatics Core Technology Platform Flow Cytometry Core Facility Histology Innovation Platform Metabolomics Innovation Resource McGill Integrated Core for Animal Modeling McGill Platform for Cellular Perturbation Single Cell Imaging and Mass Cytometry Analysis Platform.

McGill University Copyright © McGill University. WAT browning transcriptional regulation. The transcriptional regulation of the WAT beiging process involves the action of a plethora of specific proteins and nucleic acids. This orchestra of trans-acting factors modulates genes associated with oxidative capacity, mitochondrial biogenesis and non-shivering thermogenesis.

While the action of some factors, such as PGC-1α, IRF4, PRDM16, ZFP, EHMT1 and RNAs, copes with WAT browning, TLE3, ZFP, and the NuRD complex, another set of RNAs e others inhibit this process, favoring adipogenesis and white adipocyte differentiation.

Non-coding RNAs that present average length longer than nucleotides are called LncRNAs [ , ] These nucleic acids have been described to influence gene transcription through several mechanisms, including the induction of more efficient protein translation by binding to an internal ribosome entry site IRES , increase of mRNAs stability, polyubiquitination process inhibition thus increasing protein stability, binding to specific proteins in the cytoplasm, and acting as a sponge sequestering miRNAs [ , , , , ].

Zhao and collaborators identified lncRNA 1 Blnc1 as a driver for beige adipocyte differentiation and WAT browning [ ]. LncRNAs also interact with other TFs, such as PGC-1α, ZFP, and PRDM16 for full transcription [ , , ].

PRDM16, a coregulator of PPAR, cooperates with EHMT1 for WAT browning induction. As EHMT1, other covalent histone modifiers form complexes with ATP-dependent chromatin-remodelers [ ]. The chromatin-remodelers BRG1 and the BAF also interact with the TF EBF2 for gene transcription [ , ].

Noteworthy, AT functional and morphological plasticity is represented by the extreme dynamic beige adipocyte chromatin state. Beige adipocytes show a chromatin signature similar to the pattern presented by brown adipocytes after cold exposure and display a chromatin signature associated with white adipocytes after re-warming to 30 °C.

However, if the beige adipocytes were cold-induced, these cells displayed epigenetic marks that favored rapid thermogenic genes expression upon β-adrenergic stimulation, even after temperature rise, suggesting the occurrence of epigenomic memory [ , ]. For the past decades, the browning process has been deeply explored for managing both metabolic syndromes, and hypermetabolic diseases.

Proposing treatment, the cold was the pioneer mechanism associated with a great elicitation of this event in mice. However, the applicability and outcomes of this approach in humans are far from exciting.

Technological advances provide efficient mechanisms that can be employed to improve a biological system. Recent studies applied bioengineers to achieve the improvement of browning in adipose tissue. The injection of M2 AT macrophages ATMs from transgenic mwRIPKD mice, a specific knockdown of RIP that is related to activation of M1 polarization, in HFD-fed obese wild type mice could recover the disruption induced by obesity through browning induction [ ].

WAT-derived stem cells ASCs from humans and rats could be differentiated into BAT under browning conditions in three-dimensional 3D polyethylene glycol PEG hydrogel [ ].

In addition to bioengineering, a field that has been explored is the transplantation or re-implantation of BAT, also termed ex-BAT, to increase this endogenous tissue.

The harvested scWAT is differentiated into brite and then is re-implanted in the same area promoting the local increase of BAT, displaying great outcomes [ ]. Aiming for a less invasive procedure, the use of a microneedle patch to address the browning agents directly to the scWAT for inducing browning of this tissue is another strategy investigated presently [ ].

These strategies aim to promote the increase of endogenous BAT, as well as its activity with an applicable method, reducing the degree of invasiveness and risks of rejection by the body. Several studies have targeted increasing BAT mass and activity, but when the aim is inhibiting the browning process to avoid a worsening prognosis, what is proposed?

Expanding knowledge of the pathways involved is the main way of inhibiting that event. It is well established the role of parathyroid hormone PTH in the elicitation of browning.

In the tumor context, it was identified as a peptide-derived tumor, parathyroid hormone-related protein PTHrP that favors browning and cachexia [ 16 ]. Blocking PTHR specifically in ATs inhibits browning and wasting of this tissue, as well preserves muscle integrity, and ameliorates muscle-related strength, protecting mice from cachexia [ ].

Currently, some pharmacological approaches have displayed alternative and prominent ways to restrain browning. Metformin demonstrated to be efficient to prevent the browning of the scWAT, as well as the inhibition of mevalonate pathways by the use of Statin or Fluvastatin, which implicates in the disruption of browning [ , ].

However, further studies should be conducted to promote a broader and more detailed debate on the topic. Given the data presented, it is evident that the benefits and dangers associated with activation or inhibition of the browning process are closely linked to the type of disorders displayed in the individual's body at the metabolic, cellular, or physiological level.

Taking as criteria patients with metabolic syndromes and obesity, browning process emerges as a promising therapeutic approach, mainly due to its several benefits, such as improvement of clinical conditions associated with lower side effects, and the multi-stimulatory character, which allows numerous safe and feasible ways of induction.

In contrast, the ways and means of inhibiting browning to prevent the development of comorbidities in cases of chronic or acute hypermetabolism are still poorly explored.

In this way, browning process needs to be deeply explored and can be used as a key factor in different therapeutical approaches in health and diseases. Emerging insights into the metabolic and immunological role of browning of the white adipose tissue are also discussed, along with the developments that can be expected from these promising targets for therapy of metabolic and chronic disease in the forthcoming future.

A major discussion pointed by the scientific community about the investigation of AT in mice is how much the results obtained in the animal model would reproduce the anatomorphophysiologies of these tissues in humans, since, unlike humans, mice have a significant amount of BAT during embryonic and adult, as well as they differ in several other factors, such as expression of molecular markers, activation profile and location.

Human BAT was discovered to be more similar to beige compared to classical BAT markers [ , ]. Another relevant point concerns the fact that the BAT deposits most used for research purposes are the BAT located in the interscapular region iBAT , while in humans there is a greater abundance of this tissue in the clavicular and neck regions, presenting compositional differences that represent obstacles in the overlapping of scientific findings.

In this sense, Mo and colleagues identified an analogous deposit of BAT in mouse embryos, which is maintained during adulthood, and in humans called supraclavicular BAT scBAT.

The scBAT shows similarities to scBAT in humans in terms of location, morphology and thermogenic capacity [ ]. In which the authors observed a greater similarity between several cellular and molecular parameters between the BAT of humans and mice [ ]. The technique generated controversies that were discussed by Kajimura and Spiegelman and replicated by the authors of the original article , Current research place WAT browning as an extremely dynamic process that is influenced by several factors , including temperature, physical exercising, thyroid hormones, circardian rhythm, food components and dietary regimens.

The participation of AT plasticity on the organism metabolic health and inflammatory status spot this process as a promising therapeutic target for decreasing the risk associated with many chronic diseases. Further efforts in investigating AT plasticity must alleviate the burden of these devastating life-style associated pathologies.

Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. Article CAS PubMed Google Scholar. Moriya K, Arnold J, LeBlanc J. Shivering and nonshivering thermogenesis in exercised cold-deacclimated rats. Eur J Appl Physiol Occup Physiol.

Prunet-Marcassus B, Cousin B, Caton D, André M, Pénicaud L, Casteilla L. From heterogeneity to plasticity in adipose tissues: site-specific differences. Exp Cell Res. Barbatelli G, Murano I, Madsen L, Hao Q, Jimenez M, Kristiansen K, et al.

The emergence of cold-induced brown adipocytes in mouse white fat depots is determined predominantly by white to brown adipocyte transdifferentiation.

Am J Physiol Metab. CAS Google Scholar. Suárez-Zamorano N, Fabbiano S, Chevalier C, Stojanović O, Colin DJ, Stevanović A, et al. Microbiota depletion promotes browning of white adipose tissue and reduces obesity. Nat Med. Article PubMed PubMed Central CAS Google Scholar.

Johann K, Cremer AL, Fischer AW, Heine M, Pensado ER, Resch J, et al. Thyroid-hormone-induced browning of white adipose tissue does not contribute to thermogenesis and glucose consumption. Cell Rep. Barberá MJ, Schlüter A, Pedraza N, Iglesias R, Villarroya F, Giralt M. Peroxisome proliferator-activated receptor α activates transcription of the brown fat uncoupling protein-1 gene: a link between regulation of the thermogenic and lipid oxidation pathways in the brown fat cell.

J Biol Chem. Article PubMed Google Scholar. Schulz TJ, Huang TL, Tran TT, Zhang H, Townsend KL, Shadrach JL, et al. Identification of inducible brown adipocyte progenitors residing in skeletal muscle and white fat.

Proc Natl Acad Sci U S A. Villarroya F, Peyrou M, Giralt M. Transcriptional regulation of the uncoupling protein-1 gene. Kalinovich AV, de Jong JMA, Cannon B, Nedergaard J. UCP1 in adipose tissues: two steps to full browning.

Yau WW, Singh BK, Lesmana R, Zhou J, Sinha RA, Wong KA, et al. Thyroid hormone T 3 stimulates brown adipose tissue activation via mitochondrial biogenesis and MTOR-mediated mitophagy. Pérez-Martí A, Garcia-Guasch M, Tresserra-Rimbau A, Carrilho-Do-Rosário A, Estruch R, Salas-Salvadó J, et al.

A low-protein diet induces body weight loss and browning of subcutaneous white adipose tissue through enhanced expression of hepatic fibroblast growth factor 21 FGF Mol Nutr Food Res. Article CAS Google Scholar.

Finlin BS, Memetimin H, Confides AL, Kasza I, Zhu B, Vekaria HJ, et al. Human adipose beiging in response to cold and mirabegron.

JCI Insight. Article PubMed Central Google Scholar. Otero-Díaz B, Rodríguez-Flores M, Sánchez-Muñoz V, Monraz-Preciado F, Ordoñez-Ortega S, Becerril-Elias V, et al. Exercise induces white adipose tissue browning across the weight spectrum in humans. Front Physiol. Article PubMed PubMed Central Google Scholar.

Carrière A, Jeanson Y, Berger-Müller S, André M, Chenouard V, Arnaud E, et al. Browning of white adipose cells by intermediate metabolites: an adaptive mechanism to alleviate redox pressure. Article PubMed CAS Google Scholar.

Kir S, White JP, Kleiner S, Kazak L, Cohen P, Baracos VE, et al. Tumour-derived PTH-related protein triggers adipose tissue browning and cancer cachexia. Article CAS PubMed PubMed Central Google Scholar.

Abdullahi A, Samadi O, Auger C, Kanagalingam T, Boehning D, Bi S, et al. Browning of white adipose tissue after a burn injury promotes hepatic steatosis and dysfunction.

Cell Death Dis. Lucchini FC, Wueest S, Challa TD, Item F, Modica S, Borsigova M, et al. ASK1 inhibits browning of white adipose tissue in obesity. Nat Commun. Frontini A, Cinti S.

Distribution and development of brown adipocytes in the murine and human adipose organ. Cell Metab. Luo L, Liu M. Adipose tissue in control of metabolism. J Endocrinol. Kershaw EE, Flier JS. Adipose tissue as an endocrine organ.

J Clin Endocrinol Metab. Andrade-Oliveira V, Câmara NOS, Moraes-Vieira PM. Adipokines as drug targets in diabetes and underlying disturbances. J Diabetes Res. Article Google Scholar. Jokinen R, Pirnes-karhu S, Pietiläinen KH, Pirinen E. Redox Biol. Parimisetty A, Dorsemans AC, Awada R, Ravanan P, Diotel N, Lefebvre C.

Secret talk between adipose tissue and central nervous system via secreted factors-an emerging frontier in the neurodegenerative research.

J Neuroinflam. Reilly SM, Saltiel AR. Adapting to obesity with adipose tissue inflammation. Nat Rev Endocrinol. Park A. Distinction of white, beige and brown adipocytes derived from mesenchymal stem cells. World J Stem Cells.

Dalal S. Lipid metabolism in cancer cachexia. Ann Palliat Med. Wu J, Boström P, Sparks LM, Ye L, Choi JH, Giang AH, et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cuevas-Ramos D, Mehta R, Aguilar-Salinas CA. Fibroblast growth factor 21 and browning of white adipose tissue.

Bargut TCL, Souza-Mello V, Aguila MB, Mandarim-de-Lacerda CA. Browning of white adipose tissue: lessons from experimental models. Horm Mol Biol Clin Investig. Shao M, Wang QA, Song A, Vishvanath L, Busbuso NC, Scherer PE, et al.

Cellular origins of beige fat cells revisited. Vargas-Castillo A, Fuentes-Romero R, Rodriguez-Lopez LA, Torres N, Tovar AR. Understanding the biology of thermogenic fat: is browning a new approach to the treatment of obesity? Arch Med Res.

Long JZ, Svensson KJ, Tsai L, Zeng X, Roh HC, Kong X, et al. A smooth muscle-like origin for beige adipocytes. Shao M, Vishvanath L, Busbuso NC, Hepler C, Shan B, Sharma AX, et al. Oguri Y, Shinoda K, Kim H, Alba DL, Bolus WR, Wang Q, et al. CD81 controls beige fat progenitor cell growth and energy balance via FAK signaling.

Tabuchi C, Sul HS. Signaling pathways regulating thermogenesis. Front Endocrinol Lausanne. Google Scholar. Himms-hagen J, Melnyk A, Zingaretti MC, Ceresi E, Barbatelli G, Cinti S. Multilocular fat cells in WAT of CLtreated rats derive directly from white adipocytes. Am J Physiol Cell Physiol.

Golozoubova V, Cannon B, Nedergaard J. UCP1 is essential for adaptive adrenergic nonshivering thermogenesis. Am J Physiol - Endocrinol Metab. Schenk H, Franke H, Heim T. Structure and function of brown adipose tissue. Acta Histochem. Klingenberg M, Echtay KS, Bienengraeber M, Winkler E, Huang SG.

Structure-function relationship in UCP1. Int J Obes. Ricquier D, Bouillaud F. The uncoupling protein homologues: UCP1, UCP2, UCP3 StUCP and AtUCP.

Biochem J. Echtay KS. Mitochondrial uncoupling proteins—What is their physiological role? Free Radic Biol Med. Heaton JM. The distribution of brown adipose tissue in the human.

J Anat. CAS PubMed PubMed Central Google Scholar. Leitner BP, Huang S, Brychta RJ, Duckworth CJ, Baskin AS, McGehee S, et al.

Mapping of human brown adipose tissue in lean and obese young men. Proc Natl Acad Sci USA. Klingenberg M, Echtay KS. Uncoupling proteins: the issues from a biochemist point of view.

Biochim Biophys Acta Bioenerg. Chechi K, Grundberg E, Richard D, Chechi K, Vijay J, Voisine P, et al. Hoang T, Smith MD, Jelokhani-Niaraki M. Expression, folding, and proton transport activity of human uncoupling protein-1 ucp1 in lipid membranes.

Ježek P, Jabůrek M, Porter RK. Uncoupling mechanism and redox regulation of mitochondrial uncoupling protein 1 UCP1. Macher G, Koehler M, Rupprecht A, Kreiter J, Hinterdorfer P, Pohl EE.

Inhibition of mitochondrial UCP1 and UCP3 by purine nucleotides and phosphate. Biochim Biophys Acta Biomembr. Kozak UC, Kopecky J, Teisinger J, Enerbäck S, Boyer B, Kozak LP. An upstream enhancer regulating brown-fat-specific expression of the mitochondrial uncoupling protein gene.

Mol Cell Biol. Yubero P, Barbera MJ, Alvarez R, Vinas O, Mampel T, Iglesias R, et al. Dominant negative regulation by c-Jun of transcription of the uncoupling protein-1 gene through a proximal cAMP-regulatory element: a mechanism for repressing basal and norepinephrine-induced expression of the gene before brown adipocyte differentiation.

Mol Endocrinol. Manchado C, Yubero P, Vinas O, Iglesias R, Villarroya F, Mampel T, et al. Cassard-Doulcier AM, Gelly C, Bouillaud F, Ricquier D. A bp enhancer of the rat uncoupling protein-1 UCP-1 gene controls specific and regulated expression in brown adipose tissue.

Sears IB, MacGinnitie MA, Kovacs LG, Graves RA. Differentiation-dependent expression of the brown adipocyte uncoupling protein gene: regulation by peroxisome proliferator-activated receptor gamma. Wang H, Zhang Y, Yehuda-Shnaidman E, Medvedev AV, Kumar N, Daniel KW, et al.

Liver X receptor α is a transcriptional repressor of the uncoupling protein 1 gene and the brown fat phenotype. Kiskinis E, Hallberg M, Christian M, Olofsson M, Dilworth SM, White R, et al.

RIP directs histone and DNA methylation to silence Ucp1 expression in white adipocytes. EMBO J. Shore A, Karamitri A, Kemp P, Speakman JR, Lomax MA. Role of Ucp1 enhancer methylation and chromatin remodelling in the control of Ucp1 expression in murine adipose tissue.

Gebert LFR, MacRae IJ. Regulation of microRNA function in animals. Nat Rev Mol Cell Biol. Oliverio M, Schmidt E, Mauer J, Baitzel C, Hansmeier N, Khani S, et al. Dicer1-miRBace1 signalling controls brown adipose tissue differentiation and function.

Nat Cell Biol. Zhang H, Guan M, Townsend KL, Huang TL, An D, Yan X, et al. Micro RNA regulates brown adipogenesis via a novel HIF 1an- AMPK - PGC 1α signaling network. EMBO Rep. Chen SZ, Xu X, Ning LF, Jiang WY, Xing C, Tang QQ, et al.

MiR impairs the adipogenic lineage commitment via targeting lysyl oxidase. Kang T, Lu W, Xu W, Anderson L, Bacanamwo M, Thompson W, et al.

MicroRNA miR targets prohibitin and impairs adipocyte differentiation and mitochondrial function in human adipose-derived stem cells. Trajkovski M, Ahmed K, Esau CC, Stoffel M. MyomiR regulates brown fat differentiation through Prdm Enerbäck S, et al.

Mice lacking mitochondrial uncoupling protein are cold-sensitive but not obese. Liu X, Rossmeisl M, McClaine J, Kozak LP. Paradoxical resistance to diet-induced obesity in UCP1-deficient mice. J Clin Invest. Hankir MK, Seyfried F. Omran F, Christian M. Inflammatory signaling and brown fat activity.

Reynés B, van Schothorst EM, Keijer J, Ceresi E, Oliver P, Palou A. Cold induced depot-specific browning in ferret aortic perivascular adipose tissue.

Shankar K, Kumar D, Gupta S, Varshney S, Rajan S, Srivastava A, et al. Role of brown adipose tissue in modulating adipose tissue inflammation and insulin resistance in high-fat diet fed mice.

Eur J Pharmacol. van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, Drossaerts JMAFL, Kemerink GJ, Bouvy ND, et al. Cold-activated brown adipose tissue in healthy men. N Engl J Med. Feldmann HM, Golozoubova V, Cannon B, Nedergaard J. UCP1 ablation induces obesity and abolishes diet-induced thermogenesis in mice exempt from thermal stress by living at thermoneutrality.

Ohtomo T, Ino K, Miyashita R, Chigira M, Nakamura M, Someya K, et al. Chronic high-fat feeding impairs adaptive induction of mitochondrial fatty acid combustion-associated proteins in brown adipose tissue of mice. Biochem Biophys Rep. PubMed PubMed Central Google Scholar. Winn NC, Vieira-Potter VJ, Gastecki ML, Welly RJ, Scroggins RJ, Zidon TM, et al.

Loss of UCP1 exacerbates western diet-induced glycemic dysregulation independent of changes in body weight in female mice.

Am J Physiol Regul Integr Comp Physiol. Bond LM, Burhans MS, Ntambi JM. Uncoupling protein-1 deficiency promotes brown adipose tissue inflammation and ER stress.

PLoS ONE. Bond LM, Ntambi JM. UCP1 deficiency increases adipose tissue monounsaturated fatty acid synthesis and trafficking to the liver. J Lipid Res. Alcalá M, Calderon-Dominguez M, Bustos E, Ramos P, Casals N, Serra D, et al. Increased inflammation, oxidative stress and mitochondrial respiration in brown adipose tissue from obese mice.

Sci Rep. Tamucci KA, Namwanje M, Fan L, Qiang L. The dark side of browning. Protein Cell. Lim J, Park HS, Kim J, Jang YJ, Kim J-H, Lee Y, et al. Depot-specific UCP1 expression in human white adipose tissue and its association with obesity-related markers. Han J, Meng Q, Shen L, Wu G. Interleukin-6 induces fat loss in cancer cachexia by promoting white adipose tissue lipolysis and browning.

Lipids Health Dis. Petruzzelli M, Schweiger M, Schreiber R, Campos-Olivas R, Tsoli M, Allen J, et al. A switch from white to brown fat increases energy expenditure in cancer-associated cachexia.

Li L, Li B, Li M, Speakman JR. Switching on the furnace: Regulation of heat production in brown adipose tissue. Mol Asp Med. Jeschke MG. The hepatic response to thermal injury: Is the liver important for postburn outcomes? Mol Med. Cuyàs E, Verdura S, Micol V, Joven J, Bosch-Barrera J, Encinar JA, et al.

Revisiting silibinin as a novobiocin-like Hsp90 C-terminal inhibitor: computational modeling and experimental validation. Food Chem Toxicol. Cypess AM, Weiner LS, Roberts-Toler C, Elía EF, Kessler SH, Kahn PA, et al.

Activation of human brown adipose tissue by a β3-adrenergic receptor agonist. Lim S, Park J, Um JY. Front Pharmacol. Grujic D, Susulic VS, Harper M-E, Himms-Hagen J, Cunningham BA, Corkey BE, et al. Susulic VS, Frederich RC, Lawitts J, Tozzo E, Kahn BB, Harper M-E, et al. Arner P. Human fat cell lipolysis: biochemistry, regulation and clinical role.

Best Pract Res Clin Endocrinol Metab. Concha F, Prado G, Quezada J, Ramirez A, Bravo N, Flores C, et al. Nutritional and non-nutritional agents that stimulate white adipose tissue browning.

Rev Endocr Metab Disord. Seki T, Hosaka K, Lim S, Fischer C, Honek J, Yang Y, et al. Endothelial PDGF-CC regulates angiogenesis-dependent thermogenesis in beige fat.

Guilherme A, Yenilmez B, Bedard AH, Henriques F, Liu D, Lee A, et al. Control of adipocyte thermogenesis and lipogenesis through β3-adrenergic and thyroid hormone signal integration. Alcalá M, Calderon-Dominguez M, Serra D, Herrero L, Viana M.

Mechanisms of impaired brown adipose tissue recruitment in obesity. Jimenez M, Barbatelli G, Allevi R, Cinti S, Seydoux J, Giacobino JP, et al. Eur J Biochem. Herz CT, Kiefer FW.

Adipose tissue browning in mice and humans. Evans SS, Repasky EA, Fisher DT. Fever and the thermal regulation of immunity: the immune system feels the heat.

Nat Rev Immunol. Castellani JW, Young AJ. Human physiological responses to cold exposure: acute responses and acclimatization to prolonged exposure. Auton Neurosci. Gensini GF, Conti AA. J Infect. Garami A, Steiner AA, Romanovsky AA. Fever and hypothermia in systemic inflammation. Handb Clin Neurol.

Tateda K, Matsumoto T, Miyazaki S, Yamaguchi K. Lipopolysaccharide-induced lethality and cytokine production in aged mice. Infect Immun. Copeland S, Shaw Warren H, Lowry SF, Galvano SE, Remick D. Acute inflammatory response to endotoxin in mice and humans. Clin Diagn Lab Immunol.

Zhao J, Bi W, Xiao S, Lan X, Cheng X, Zhang J, et al. Neuroinflammation induced by lipopolysaccharide causes cognitive impairment in mice. Rudaya AY, Steiner AA, Robbins JR, Dragic AS, Romanovsky AA. Thermoregulatory responses to lipopolysaccharide in the mouse: dependence on the dose and ambient temperature.

Corrigan JJ, Fonseca MT, Flatow EA, Lewis K, Steiner AA. Hypometabolism and hypothermia in the rat model of endotoxic shock: Independence of circulatory hypoxia. J Physiol. Wanner SP, Yoshida K, Kulchitsky VA, Ivanov AI, Kanosue K, Romanovsky AA.

Lipopolysaccharide-induced neuronal activation in the paraventricular and dorsomedial hypothalamus depends on ambient temperature. Osaka T. Lipopolysaccharide-induced thermogenesis mediated by GABA in the preoptic area of anesthetized rats. J Therm Biol. Tham S, Thompson R, Landeg O, Murray KA, Waite T.

Indoor temperature and health: a global systematic review. Public Health. Cheshire WP. Thermoregulatory disorders and illness related to heat and cold stress. Rumbus Z, Matics R, Hegyi P, Zsiboras C, Szabo I, Illes A, et al. Fever is associated with reduced, hypothermia with increased mortality in septic patients: a meta-analysis of clinical trials.

Fu SH, Gasparrini A, Rodriguez PS, Jha P. Mortality attributable to hot and cold ambient temperatures in India: a nationally representative case-crossover study. PLoS Med. Chen R, Yin P, Wang L, Liu C, Niu Y, Wang W, et al.

Association between ambient temperature and mortality risk and burden: time series study in main Chinese cities. Lee SL, Battistella FD, Go K. Hypothermia induces T-cell production of immunosuppressive cytokines.

J Surg Res. Kurz A, Sessler DI, Lenhardt R. Perioperative normothermia to reduce the incidence of surgical-wound infection and shorten hospitalization. Hylander BL, Repasky EA. Thermoneutrality, mice, and cancer: a heated opinion. Trends Cancer. Mace TA, Zhong L, Kilpatrick C, Zynda E, Lee C-T, Capitano M, et al.

J Leukoc Biol. Kokolus KM, Capitano ML, Lee CT, Eng JWL, Waight JD, Hylander BL, et al. Baseline tumor growth and immune control in laboratory mice are significantly influenced by subthermoneutral housing temperature.

Du G, Liu Y, Li J, Liu W, Wang Y, Li H. Hypothermic microenvironment plays a key role in tumor immune subversion. Int Immunopharmacol. Dieing A, Ahlers O, Hildebrandt B, Kerner T, Tamm I, Possinger K, et al. The effect of induced hyperthermia on the immune system. Prog Brain Res. Kobayashi Y, Ito Y, Ostapenko VV, Sakai M, Matsushita N, Imai K, et al.

Fever-range whole-body heat treatment stimulates antigen-specific T-cell responses in humans. Immunol Lett. Ostberg JR, Dayanc BE, Yuan M, Oflazoglu E, Repasky EA. Enhancement of natural killer NK cell cytotoxicity by fever-range thermal stress is dependent on NKG2D function and is associated with plasma membrane NKG2D clustering and increased expression of MICA on target cells.

Umar D, Das A, Gupta S, Chattopadhyay S, Sarkar D, Mirji G, et al. Febrile temperature change modulates CD4 T cell differentiation via a TRPV channel-regulated Notch-dependent pathway. Matsumoto K, Yamamoto N, Hagiwara S, Saito M, Furue H, Shigetomi T, et al.

Optimization of hyperthermia and dendritic cell immunotherapy for squamous cell carcinoma. Oncol Rep. PubMed Google Scholar. Jackson TC, Kochanek PM. A new vision for therapeutic hypothermia in the era of targeted temperature management: a speculative synthesis.

Ther Hypotherm Temp Manag. Gu LJ, Xiong XX, Ito T, Lee J, Xu BH, Krams S, et al. Moderate hypothermia inhibits brain inflammation and attenuates stroke-induced immunodepression in rats.

CNS Neurosci Ther. Stanek A, Cholewka A, Wielkoszyński T, Romuk E, Sieroń A. Whole-body cryotherapy decreases the levels of inflammatory, oxidative stress, and atherosclerosis plaque markers in male patients with active-phase ankylosing spondylitis in the absence of classical cardiovascular risk factors.

Mediators Inflamm. Lee JH, Wei ZZ, Cao W, Won S, Gu X, Winter M, et al. Regulation of therapeutic hypothermia on inflammatory cytokines, microglia polarization, migration and functional recovery after ischemic stroke in mice.

Neurobiol Dis. Wilson LJ, Dimitriou L, Hills FA, Gondek MB, Cockburn E. Whole body cryotherapy, cold water immersion, or a placebo following resistance exercise: a case of mind over matter? Eur J Appl Physiol. Ziemann E, Olek RA, Grzywacz T, Kaczor JJ, Antosiewicz J, Skrobot W, et al.

Whole-body cryostimulation as an effective way of reducing exercise-induced inflammation and blood cholesterol in young men. Eur Cytokine Netw. Stanek A, Romuk E, Wielkoszyński T, Bartuś S, Cieślar G, Cholewka A. Decreased lipid profile and oxidative stress in healthy subjects who underwent whole-body cryotherapy in closed cryochamber with subsequent kinesiotherapy.

Oxid Med Cell Longev. Coolbaugh CL, Damon BM, Bush EC, Welch EB, Towse TF. Cold exposure induces dynamic, heterogeneous alterations in human brown adipose tissue lipid content.

Sun YJ, Zhang ZY, Fan B, Li GY. Neuroprotection by therapeutic hypothermia. Front Neurosci. Svedung Wettervik TM, Engquist H, Lenell S, Howells T, Hillered L, Rostami E, et al. Systemic hyperthermia in traumatic brain injury—relation to intracranial pressure dynamics, cerebral energy metabolism, and clinical outcome.

J Neurosurg Anesthesiol. Bain AR, Hoiland RL, Donnelly J, Nowak-Flück D, Sekhon M, Tymko MM, et al. Cerebral metabolism, oxidation and inflammation in severe passive hyperthermia with and without respiratory alkalosis. Thorne AM, Ubbink R, Bruggenwirth IMA, Nijsten MW, Porte RJ, de Meijer VE. Hyperthermia-induced changes in liver physiology and metabolism: a rationale for hyperthermic machine perfusion.

Am J Physiol Gastrointest Liver Physiol. Clapham JC. Central control of thermogenesis. Morrison SF. Central neural control of thermoregulation and brown adipose tissue.

Fan G, Dang X, Li Y, Chen J, Zhao R, Yang X. Zinc-α2-glycoprotein promotes browning of white adipose tissue in cold-exposed male mice. Mol Cell Endocrinol. Luo X, Jia R, Zhang Q, Sun B, Yan J.

Cold-Induced browning dynamically alters the expression profiles of inflammatory adipokines with tissue specificity in mice. Int J Mol Sci. Article PubMed Central CAS Google Scholar.

Reitman ML. Of mice and men—environmental temperature, body temperature, and treatment of obesity. FEBS Lett. Brychta RJ, Huang S, Wang J, Leitner BP, Hattenbach JD, Bell SL, et al.

Quantification of the capacity for cold-induced thermogenesis in young men with and without obesity. Blauw LL, Aziz NA, Tannemaat MR, Blauw CA, de Craen AJ, Pijl H, et al. Diabetes incidence and glucose intolerance prevalence increase with higher outdoor temperature.

BMJ Open Diabetes Res Care. Kanazawa S. Does global warming contribute to the obesity epidemic? Environ Res.

Li Y, Wang D, Ping X, Zhang Y, Zhang T, Wang L, et al. Local hyperthermia therapy induces browning of white fat and treats obesity. Patsouris D, Qi P, Abdullahi A, Stanojcic M, Chen P, Parousis A, et al.

Burn induces browning of the subcutaneous white adipose tissue in mice and humans. Vinaik R, Barayan D, Abdullahi A, Jeschke MG. NLRP3 inflammasome mediates white adipose tissue browning after burn.

Am J Physiol Endocrinol Metab. Böstrom P, Wu JJM, et al. A PGC1- a -dependent myokine that drives brown-fat-like development of white fat and thermogenesis. β-Aminoisobutyric acid induces browning of white fat and hepatic β-oxidation and is inversely correlated with cardiometabolic risk factors.

Cao W, Daniel KW, Robidoux J, Puigserver P, Medvedev AV, Bai X, et al. p38 Mitogen-activated protein kinase is the central regulator of cyclic AMP-dependent transcription of the brown fat uncoupling protein 1 gene. Powers SK, Deminice R, Ozdemir M, Yoshihara T, Bomkamp MP, Hyatt H.

Research Tossue Cell biology Free Antioxidant-rich foods for brain health Adipoe correspondence to: Ulf Smith, Lundberg Laboratory for Diabetes Tizsue, Department of Molecular and Clinical Medicine, Sahlgrenska Academy at the University of Gothenburg, Blå Stråket 5, Gothenburg, Sweden. Phone: ; Email: ulf. smith medic. Find articles by Gogg, S. in: JCI PubMed Google Scholar. Find articles by Nerstedt, A. Thank you Digestive health benefits explained visiting Well-regulated adipose tissue. Well-regulate are using a browser version with limited support adiposw CSS. To obtain the best experience, we recommend you use a Well-regulated adipose tissue adippose to date browser or turn off compatibility mode in Internet Explorer. In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript. Brown and beige adipose tissue are emerging as distinct endocrine organs. These tissues are functionally associated with skeletal muscle, adipose tissue metabolism and systemic energy expenditure, suggesting an interorgan signaling network.

Author: Akinokazahn

5 thoughts on “Well-regulated adipose tissue

  1. Ich tue Abbitte, dass sich eingemischt hat... Ich finde mich dieser Frage zurecht. Man kann besprechen.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com