1
|
Cypess AM, Cannon B, Nedergaard J, Kazak L, Chang DC, Krakoff J, Tseng YH, Schéele C, Boucher J, Petrovic N, Blondin DP, Carpentier AC, Virtanen KA, Kooijman S, Rensen PCN, Cero C, Kajimura S. Emerging debates and resolutions in brown adipose tissue research. Cell Metab 2025; 37:12-33. [PMID: 39644896 PMCID: PMC11710994 DOI: 10.1016/j.cmet.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/28/2024] [Accepted: 11/01/2024] [Indexed: 12/09/2024]
Abstract
Until two decades ago, brown adipose tissue (BAT) was studied primarily as a thermogenic organ of small rodents in the context of cold adaptation. The discovery of functional human BAT has opened new opportunities to understand its physiological role in energy balance and therapeutic applications for metabolic disorders. Significantly, the role of BAT extends far beyond thermogenesis, including glucose and lipid homeostasis, by releasing mediators that communicate with other cells and organs. The field has made major advances by using new model systems, ranging from subcellular studies to clinical trials, which have also led to debates. In this perspective, we identify six fundamental issues that are currently controversial and comprise dichotomous models. Each side presents supporting evidence and, critically, the necessary methods and falsifiable experiments that would resolve the dispute. With this collaborative approach, the field will continue to productively advance the understanding of BAT physiology, appreciate the importance of thermogenic adipocytes as a central area of ongoing research, and realize the therapeutic potential.
Collapse
Affiliation(s)
- Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Douglas C Chang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Camilla Schéele
- Novo Nordisk Foundation Center for Basic Metabolic Research, The Center of Inflammation and Metabolism and the Center for Physical Activity Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Natasa Petrovic
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Cheryl Cero
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
2
|
Predescu DN, Mokhlesi B, Predescu SA. X-inactive-specific transcript: a long noncoding RNA with a complex role in sex differences in human disease. Biol Sex Differ 2024; 15:101. [PMID: 39639337 PMCID: PMC11619133 DOI: 10.1186/s13293-024-00681-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
In humans, the X and Y chromosomes determine the biological sex, XX specifying for females and XY for males. The long noncoding RNA X-inactive specific transcript (lncRNA XIST) plays a crucial role in the process of X chromosome inactivation (XCI) in cells of the female, a process that ensures the balanced expression of X-linked genes between sexes. Initially, it was believed that XIST can be expressed only from the inactive X chromosome (Xi) and is considered a typically female-specific transcript. However, accumulating evidence suggests that XIST can be detected in male cells as well, and it participates in the development of cancers and other human diseases by regulating gene expression at epigenetic, chromatin remodeling, transcriptional, and translational levels. XIST is abnormally expressed in many sexually dimorphic diseases, including autoimmune and neurological diseases, pulmonary arterial hypertension (PAH), and some types of cancers. However, the underlying mechanisms are not fully understood. Escape from XCI and skewed XCI also contributes to sex-biased diseases and their severity. Interestingly, in humans, similar to experimental animal models of human disease, the males with the XIST gene activated display the sex-biased disease condition at a rate close to females, and significantly greater than males who had not been genetically modified. For instance, the men with supernumerary X chromosomes, such as men with Klinefelter syndrome (47, XXY), are predisposed toward autoimmunity similar to females (46, XX), and have increased risk for strongly female biased diseases, compared to 46, XY males. Interestingly, chromosome X content has been linked to a longer life span, and the presence of two chromosome X contributes to increased longevity regardless of the hormonal status. In this review, we summarize recent knowledge about XIST structure/function correlation and involvement in human disease with focus on XIST abnormal expression in males. Many human diseases show differences between males and females in penetrance, presentation, progression, and survival. In humans, the X and Y sex chromosomes determine the biological sex, XX specifying for females and XY for males. This numeric imbalance, two X chromosomes in females and only one in males, known as sex chromosome dosage inequality, is corrected in the first days of embryonic development by inactivating one of the X chromosomes in females. While this "dosage compensation" should in theory solve the difference in the number of genes between sexes, the expressed doses of X genes are incompletely compensated by X chromosome inactivation in females. In this review we try to highlight how abnormal expression and function of XIST, a gene on the X chromosome responsible for this inactivation process, may explain the sex differences in human health and disease. A better understanding of the molecular mechanisms of XIST participation in the male-female differences in disease is highly relevant since it would allow for improving the personalization of diagnosis and sex-specific treatment of patients.
Collapse
Affiliation(s)
- Dan N Predescu
- Department of Internal Medicine, Pulmonary, Critical Care, and Sleep Medicine, Rush University Medical Center, Chicago, IL, 60612, USA.
| | - Babak Mokhlesi
- Department of Internal Medicine, Pulmonary, Critical Care, and Sleep Medicine, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Sanda A Predescu
- Department of Internal Medicine, Pulmonary, Critical Care, and Sleep Medicine, Rush University Medical Center, Chicago, IL, 60612, USA
| |
Collapse
|
3
|
Lambert J, Kovilakath A, Jamil M, Valentine Y, Anderson A, Montefusco D, Cowart LA. Sphingosine kinase 1 is induced by glucocorticoids in adipose derived stem cells and enhances glucocorticoid mediated signaling in adipose expansion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612482. [PMID: 39314417 PMCID: PMC11419133 DOI: 10.1101/2024.09.13.612482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Sphingosine kinase 1 (SphK1) plays a crucial role in regulating metabolic pathways within adipocytes and is elevated in the adipose tissue of obese mice. While previous studies have reported both pro- and inhibitory effects of SphK1 and its product, sphingosine-1-phosphate (S1P), on adipogenesis, the precise mechanisms remain unclear. This study explores the timing and downstream effects of SphK1/S1P expression and activation during in vitro adipogenesis. We demonstrate that the synthetic glucocorticoid dexamethasone robustly induces SphK1 expression, suggesting its involvement in glucocorticoid-dependent signaling during adipogenesis. Notably, the activation of C/EBPδ, a key gene in early adipogenesis and a target of glucocorticoids, is diminished in SphK1-/- adipose-derived stem cells (ADSCs). Furthermore, glucocorticoid administration promotes adipose tissue expansion via SphK1 in a depot-specific manner. Although adipose expansion still occurs in SphK1-/- mice, it is significantly reduced. These findings indicate that while SphK1 is not essential for adipogenesis, it enhances early gene activation, thereby facilitating adipose tissue expansion.
Collapse
Affiliation(s)
- Johana Lambert
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
- Departments of Biochemistry and Molecular Biology Medical University of South Carolina, Charleston, SC, USA
| | - Anna Kovilakath
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Maryam Jamil
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Yolander Valentine
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrea Anderson
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
- Departments of Biochemistry and Molecular Biology Medical University of South Carolina, Charleston, SC, USA
| | - David Montefusco
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
- Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, USA
| |
Collapse
|
4
|
Bolin AP, de Fatima Silva F, Salgueiro RB, Dos Santos BA, Komino ACM, Andreotti S, de Sousa É, de Castro É, Real CC, de Paula Faria D, Souza GP, Camara H, Sorgi CA, Tseng YH, Lima FB, Rodrigues AC. Glucocorticoid modulates oxidative and thermogenic function of rat brown adipose tissue and human brown adipocytes. J Cell Physiol 2024; 239:1-12. [PMID: 39091018 DOI: 10.1002/jcp.31397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/09/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
Chronic and excessive glucocorticoid (GC) exposure can cause Cushing's syndrome, resulting in fat accumulation in selected body areas. Particularly in the brown adipose tissue (BAT), GC acts negatively, resulting in whitening of the tissue. We hypothesized that dysregulation of microRNAs by GC could be an additional mechanism to explain its negative actions in BAT. Male Wistar rats were divided into two groups: (1) Control sham and (2) GC group that was administered dexamethasone 6.25 mg/200 μL via osmotic pump implantation over 28 days. After this period, the animals were euthanized and BAT tissue was properly stored. Human fat cells treated with dexamethasone were used to translate the experimental results found in animals to human biology. GC-treated rat BAT presented with large lipid droplets, severely impaired thermogenic activation, and reduced glucose uptake measured by 18F-FDG PET/CT. GC exposure induced a reduction in the mitochondrial OXPHOS system and oxygen consumption. MicroRNA profiling of BAT revealed five top-regulated microRNAs and among them miR-21-5p was the most significantly upregulated in GC-treated rats compared to the control group. Although upregulation of miR-21-5p in the tissue, differentiated primary brown adipocytes from GC-treated rats had decreased miR-21-5p levels compared to the control group. To translate these results to the clinic, human brown adipocytes were treated with dexamethasone and miR-21-5p inhibitor. In human brown cells, inhibition of miR-21-5p increased brown adipocyte differentiation and prevented GC-induced glucose uptake, resulting in a lower glycolysis rate. In conclusion, high-dose GC therapy significantly impacts brown adipose tissue function, with a notable association between glucose uptake and miR-21-5p.
Collapse
Affiliation(s)
- Anaysa Paola Bolin
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Flaviane de Fatima Silva
- Department of Physiology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Rafael Barrera Salgueiro
- Department of Physiology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Bruna Araújo Dos Santos
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Sandra Andreotti
- Department of Physiology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Érica de Sousa
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Érique de Castro
- Department of Physiology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Caroline Cristiano Real
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Daniele de Paula Faria
- Department of Radiology and Oncology, Laboratory of Nuclear Medicine (LIM43), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Gerson Profeta Souza
- Department of Medicine, Section on Integrative Physiology and Metabolism, Joslin Diabetes Center Harvard Medical School, Boston, Massachusetts, USA
| | - Henrique Camara
- Department of Medicine, Section on Integrative Physiology and Metabolism, Joslin Diabetes Center Harvard Medical School, Boston, Massachusetts, USA
| | - Carlos Arterio Sorgi
- Department of Biochemistry and Immunology, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Ribeirão Preto, Brazil
- Department of Chemistry, Faculdade de Filosofia, Ciencias e Letras de Ribeirão Preto - FFCLRP/USP, Ribeirão Preto, Brazil
| | - Yu-Hua Tseng
- Department of Medicine, Section on Integrative Physiology and Metabolism, Joslin Diabetes Center Harvard Medical School, Boston, Massachusetts, USA
| | - Fábio Bessa Lima
- Department of Physiology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Alice Cristina Rodrigues
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
5
|
Prapaharan B, Lea M, Beaudry JL. Weighing in on the role of brown adipose tissue for treatment of obesity. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13157. [PMID: 39087083 PMCID: PMC11290130 DOI: 10.3389/jpps.2024.13157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024]
Abstract
Brown adipose tissue (BAT) activation is an emerging target for obesity treatments due to its thermogenic properties stemming from its ability to shuttle energy through uncoupling protein 1 (Ucp1). Recent rodent studies show how BAT and white adipose tissue (WAT) activity can be modulated to increase the expression of thermogenic proteins. Consequently, these alterations enable organisms to endure cold-temperatures and elevate energy expenditure, thereby promoting weight loss. In humans, BAT is less abundant in obese subjects and impacts of thermogenesis are less pronounced, bringing into question whether energy expending properties of BAT seen in rodents can be translated to human models. Our review will discuss pharmacological, hormonal, bioactive, sex-specific and environmental activators and inhibitors of BAT to determine the potential for BAT to act as a therapeutic strategy. We aim to address the feasibility of utilizing BAT modulators for weight reduction in obese individuals, as recent studies suggest that BAT's contributions to energy expenditure along with Ucp1-dependent and -independent pathways may or may not rectify energy imbalance characteristic of obesity.
Collapse
Affiliation(s)
| | | | - Jacqueline L. Beaudry
- Temerty Faculty of Medicine, Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
6
|
Riera CE. Wiring the Brain for Wellness: Sensory Integration in Feeding and Thermogenesis: A Report on Research Supported by Pathway to Stop Diabetes. Diabetes 2024; 73:338-347. [PMID: 38377445 PMCID: PMC10882152 DOI: 10.2337/db23-0706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/06/2023] [Indexed: 02/22/2024]
Abstract
The recognition of sensory signals from within the body (interoceptive) and from the external environment (exteroceptive), along with the integration of these cues by the central nervous system, plays a crucial role in maintaining metabolic balance. This orchestration is vital for regulating processes related to both food intake and energy expenditure. Animal model studies indicate that manipulating specific populations of neurons in the central nervous system which influence these processes can effectively modify energy balance. This body of work presents an opportunity for the development of innovative weight loss therapies for the treatment of obesity and type 2 diabetes. In this overview, we delve into the sensory cues and the neuronal populations responsible for their integration, exploring their potential in the development of weight loss treatments for obesity and type 2 diabetes. This article is the first in a series of Perspectives that report on research funded by the American Diabetes Association Pathway to Stop Diabetes program. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Céline E. Riera
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
7
|
Anekwe CV, Ahn YJ, Bajaj SS, Stanford FC. Pharmacotherapy causing weight gain and metabolic alteration in those with obesity and obesity-related conditions: A review. Ann N Y Acad Sci 2024; 1533:145-155. [PMID: 38385953 PMCID: PMC11057385 DOI: 10.1111/nyas.15112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
This review aims to summarize pharmacological interventions that may affect adiposity and metabolic equilibrium in individuals with obesity. Pharmacological therapy is frequently used to treat medical conditions that are both directly related to obesity (such as hypertension and type 2 diabetes) and indirectly related to obesity (such as asthma, insomnia, and type 1 diabetes). This pharmacological therapy may result in weight gain and alterations in the metabolic profile. Many medication classes are implicated in the pharmacologic causes of weight gain, including antipsychotics, glucocorticoids, beta-adrenergic blockers, tricyclic antidepressants, antihistamines, insulin, neuropathic agents, sleep agents, and steroids. This article describes the mechanisms of action and pathways of pharmacological interventions causing obesity.
Collapse
Affiliation(s)
- Chika V. Anekwe
- Massachusetts General Hospital, MGH Weight Center, Department of Internal Medicine-Division of Endocrinology-Metabolism Unit, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yoon Ji Ahn
- Massachusetts General Hospital, MGH Weight Center, Department of Internal Medicine-Division of Endocrinology-Metabolism Unit, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Fatima Cody Stanford
- Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital, MGH Weight Center, Department of Internal Medicine-Division of Endocrinology-Neuroendocrine Unit and Department of Pediatrics-Division of Pediatric Endocrinology, Nutrition Obesity Research Center at Harvard (NORCH), Boston, MA, USA
| |
Collapse
|
8
|
Giordano AP, Gambaro SE, Alzamendi A, Harnichar AE, Rey MA, Ongaro L, Spinedi E, Zubiría MG, Giovambattista A. Dexamethasone Inhibits White Adipose Tissue Browning. Int J Mol Sci 2024; 25:2714. [PMID: 38473960 DOI: 10.3390/ijms25052714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/20/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
White adipose tissue (WAT) regulates energy balance through energy storage, adipokines secretion and the thermogenesis process. Beige adipocytes are responsible for WAT thermogenesis. They are generated by adipogenesis or transdifferentiation during cold or β3-adrenergic agonist stimulus through a process called browning. Browning has gained significant interest for to its preventive effect on obesity. Glucocorticoids (GCs) have several functions in WAT biology; however, their role in beige adipocyte generation and WAT browning is not fully understood. The aim of our study was to determine the effect of dexamethasone (DXM) on WAT thermogenesis. For this purpose, rats were treated with DXM at room temperature (RT) or cold conditions to determine different thermogenic markers. Furthermore, the effects of DXM on the adipogenic potential of beige precursors and on mature beige adipocytes were evaluated in vitro. Our results showed that DXM decreased UCP-1 mRNA and protein levels, mainly after cold exposure. In vitro studies showed that DXM decreased the expression of a beige precursor marker (Ebf2), affecting their ability to differentiate into beige adipocytes, and inhibited the thermogenic response of mature beige adipocytes (Ucp-1, Dio2 and Pgc1α gene expressions and mitochondrial respiration). Overall, our data strongly suggest that DXM can inhibit the thermogenic program of both retroperitoneal and inguinal WAT depots, an effect that could be exerted, at least partially, by inhibiting de novo cell generation and the thermogenic response in beige adipocytes.
Collapse
Affiliation(s)
- Alejandra Paula Giordano
- Neuroendocrinology Laboratory, Multidisciplinary Institute of Cellular Biology (IMBICE, CICPBA-CONICET-UNLP), La Plata 1900, Argentina
- Biology Department, School of Exact Sciences, Universidad Nacional de La Plata, La Plata 1900, Argentina
| | - Sabrina Eliana Gambaro
- Neuroendocrinology Laboratory, Multidisciplinary Institute of Cellular Biology (IMBICE, CICPBA-CONICET-UNLP), La Plata 1900, Argentina
- Biology Department, School of Exact Sciences, Universidad Nacional de La Plata, La Plata 1900, Argentina
| | - Ana Alzamendi
- Neuroendocrinology Laboratory, Multidisciplinary Institute of Cellular Biology (IMBICE, CICPBA-CONICET-UNLP), La Plata 1900, Argentina
| | - Alejandro Ezequiel Harnichar
- Neuroendocrinology Laboratory, Multidisciplinary Institute of Cellular Biology (IMBICE, CICPBA-CONICET-UNLP), La Plata 1900, Argentina
| | - María Amanda Rey
- Neuroendocrinology Laboratory, Multidisciplinary Institute of Cellular Biology (IMBICE, CICPBA-CONICET-UNLP), La Plata 1900, Argentina
| | - Luisina Ongaro
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 0G4, Canada
| | - Eduardo Spinedi
- CENEXA (UNLP-CONICET), La Plata Medical School-UNLP, Calles 60 y 120, La Plata 1900, Argentina
| | - María Guillermina Zubiría
- Neuroendocrinology Laboratory, Multidisciplinary Institute of Cellular Biology (IMBICE, CICPBA-CONICET-UNLP), La Plata 1900, Argentina
- Biology Department, School of Exact Sciences, Universidad Nacional de La Plata, La Plata 1900, Argentina
| | - Andrés Giovambattista
- Neuroendocrinology Laboratory, Multidisciplinary Institute of Cellular Biology (IMBICE, CICPBA-CONICET-UNLP), La Plata 1900, Argentina
- Biology Department, School of Exact Sciences, Universidad Nacional de La Plata, La Plata 1900, Argentina
| |
Collapse
|
9
|
Zhang Y, Du C, Wang W, Qiao W, Li Y, Zhang Y, Sheng S, Zhou X, Zhang L, Fan H, Yu Y, Chen Y, Liao Y, Chen S, Chang Y. Glucocorticoids increase adiposity by stimulating Krüppel-like factor 9 expression in macrophages. Nat Commun 2024; 15:1190. [PMID: 38331933 PMCID: PMC10853261 DOI: 10.1038/s41467-024-45477-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
The mechanisms underlying glucocorticoid (GC)-induced obesity are poorly understood. Macrophages are the primary targets by which GCs exert pharmacological effects and perform critical functions in adipose tissue homeostasis. Here, we show that macrophages are essential for GC-induced obesity. Dexamethasone (Dex) strongly induced Krüppel-like factor 9 (Klf9) expression in macrophages. Similar to Dex, lentivirus-mediated Klf9 overexpression inhibits M1 and M2a markers expression, causing macrophage deactivation. Furthermore, the myeloid-specific Klf9 transgene promotes obesity. Conversely, myeloid-specific Klf9-knockout (mKlf9KO) mice are lean. Moreover, myeloid Klf9 knockout largely blocks obesity induced by chronic GC treatment. Mechanistically, GC-inducible KLF9 recruits the SIN3A/HDAC complex to the promoter regions of Il6, Ptgs2, Il10, Arg1, and Chil3 to inhibit their expression, subsequently reducing thermogenesis and increasing lipid accumulation by inhibiting STAT3 signaling in adipocytes. Thus, KLF9 in macrophages integrates the beneficial anti-inflammatory and adverse metabolic effects of GCs and represents a potential target for therapeutic interventions.
Collapse
Affiliation(s)
- Yinliang Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Chunyuan Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Wei Wang
- Key Laboratory of Biotechnology of Hubei Province, Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei University, Wuhan, China
| | - Wei Qiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Yuhui Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Yujie Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Sufang Sheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Xuenan Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Lei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Heng Fan
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, Ningxia, China
| | - Ying Yu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yong Chen
- Key Laboratory of Biotechnology of Hubei Province, Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei University, Wuhan, China
| | - Yunfei Liao
- Department of Endocrinology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shihong Chen
- Department of Endocrinology, The Second Hospital of Shandong University, Jinan, China.
| | - Yongsheng Chang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
10
|
Kugler BA, Cao X, Wenger M, Franczak E, McCoin CS, Von Schulze A, Morris EM, Thyfault JP. Divergence in aerobic capacity influences hepatic and systemic metabolic adaptations to bile acid sequestrant and short-term high-fat/sucrose feeding in rats. Am J Physiol Regul Integr Comp Physiol 2023; 325:R712-R724. [PMID: 37811712 PMCID: PMC11178297 DOI: 10.1152/ajpregu.00133.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
High versus low aerobic capacity significantly impacts the risk for metabolic diseases. Rats selectively bred for high or low intrinsic aerobic capacity differently modify hepatic bile acid metabolism in response to high-fat diets (HFDs). Here we tested if a bile acid sequestrant would alter hepatic and whole body metabolism differently in rats with high and low aerobic capacity fed a 1-wk HFD. Male rats (8 mo of age) that were artificially selected to be high (HCR) and low-capacity runners (LCR) with divergent intrinsic aerobic capacities were transitioned from a low-fat diet (LFD, 10% fat) to an HFD (45% fat) with or without a bile acid sequestrant (BA-Seq, 2% cholestyramine resin) for 7 days while maintained in an indirect calorimetry system. HFD + BA-Seq increased fecal excretion of lipids and bile acids and prevented weight and fat mass gain in both strains. Interestingly, HCR rats had increased adaptability to enhance fecal bile acid and lipid loss, resulting in more significant energy loss than their LCR counterpart. In addition, BA-Seq induced a greater expression of hepatic CYP7A1 gene expression, the rate-limiting enzyme of bile acid synthesis in HCR rats both on HFD and HFD + BA-Seq diets. HCR displayed a more significant reduction of RQ in response to HFD than LCR, but HFD + BA-Seq lowered RQ in both groups compared with HFD alone, demonstrating a pronounced impact on metabolic flexibility. In conclusion, BA-Seq provides uniform metabolic benefits for metabolic flexibility and adiposity, but rats with higher aerobic capacity display adaptability for hepatic bile acid metabolism.NEW & NOTEWORTHY The administration of bile acid sequestrant (BA-Seq) has uniform metabolic benefits in terms of metabolic flexibility and adiposity in rats with high and low aerobic capacity. However, rats with higher aerobic capacity demonstrate greater adaptability in hepatic bile acid metabolism, resulting in increased fecal bile acid and lipid loss, as well as enhanced fecal energy loss.
Collapse
Affiliation(s)
- Benjamin A Kugler
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Xin Cao
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Madi Wenger
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Edziu Franczak
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| | - Colin S McCoin
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| | - Alex Von Schulze
- Stowers Research Institute, Kansas City, Missouri, United States
| | - E Matthew Morris
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - John P Thyfault
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| |
Collapse
|
11
|
Zhang W, Nowotny H, Theodoropoulou M, Simon J, Hemmer CM, Bidlingmaier M, Auer MK, Reincke M, Uhlenhaut H, Reisch N. E47 as a novel glucocorticoid-dependent gene mediating lipid metabolism in patients with endogenous glucocorticoid excess. Front Endocrinol (Lausanne) 2023; 14:1249863. [PMID: 38047107 PMCID: PMC10691538 DOI: 10.3389/fendo.2023.1249863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/26/2023] [Indexed: 12/05/2023] Open
Abstract
Purpose E47 has been identified as a modulating transcription factor of glucocorticoid receptor target genes, its loss protecting mice from metabolic adverse effects of glucocorticoids. We aimed to analyze the role of E47 in patients with endogenous glucocorticoid excess [Cushing's syndrome (CS)] and its association with disorders of lipid and glucose metabolism. Methods This is a prospective cohort study including 120 female patients with CS (ACTH-dependent = 79; ACTH-independent = 41) and 26 healthy female controls. Morning whole blood samples after an overnight fast were used to determine E47 mRNA expression levels in patients with overt CS before and 6-12 months after curative surgery. Expression levels were correlated with the clinical phenotype of the patients. Control subjects underwent ACTH stimulation tests and dexamethasone suppression tests to analyze short-term regulation of E47. Results E47 gene expression showed significant differences in patient cohorts with overt CS vs. patients in remission (p = 0.0474) and in direct intraindividual comparisons pre- vs. post-surgery (p = 0.0353). ACTH stimulation of controls resulted in a significant decrease of E47 mRNA expression 30 min after i.v. injection compared to baseline measurements. Administration of 1 mg of dexamethasone overnight in controls did not change E47 mRNA expression. E47 gene expression showed a positive correlation with total serum cholesterol (p = 0.0036), low-density lipoprotein cholesterol (p = 0.0157), and waist-arm ratio (p = 0.0138) in patients with CS in remission. Conclusion E47 is a GC-dependent gene that is upregulated in GC excess potentially aiming at reducing metabolic glucocorticoid side effects such as dyslipidemia.
Collapse
Affiliation(s)
- Wei Zhang
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Hanna Nowotny
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Marily Theodoropoulou
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Julia Simon
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Charlotte M. Hemmer
- Molecular Endocrinology, Institutes for Diabetes and Obesity & Diabetes and Cancer IDO & IDC, Helmholtz Zentrum Muenchen (HMGU) and German Center for Diabetes Research (DZD), Munich, Germany
| | - Martin Bidlingmaier
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Matthias K. Auer
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Martin Reincke
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Henriette Uhlenhaut
- Molecular Endocrinology, Institutes for Diabetes and Obesity & Diabetes and Cancer IDO & IDC, Helmholtz Zentrum Muenchen (HMGU) and German Center for Diabetes Research (DZD), Munich, Germany
- Metabolic Programming, Technische Universität München (TUM) School of Life Sciences Weihenstephan and ZIEL Institute for Food & Health, Munich, Germany
| | - Nicole Reisch
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| |
Collapse
|
12
|
Opałka B, Żołnierczuk M, Grabowska M. Immunosuppressive Agents-Effects on the Cardiovascular System and Selected Metabolic Aspects: A Review. J Clin Med 2023; 12:6935. [PMID: 37959400 PMCID: PMC10647341 DOI: 10.3390/jcm12216935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 10/28/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
The widespread use of immunosuppressive drugs makes it possible to reduce inflammation in autoimmune diseases, as well as prevent transplant rejection in organ recipients. Despite their key action in blocking the body's immune response, these drugs have many side effects. These actions primarily affect the cardiovascular system, and the incidence of complications in patients using immunosuppressive drugs is significant, being associated with a higher incidence of cardiovascular incidents such as myocardial infarction and stroke. This paper analyzes the mechanisms of action of commonly used immunosuppressive drugs and their impact on the cardiovascular system. The adverse effect of immunosuppressive drugs is associated with toxicity within the cardiovascular system, which may be a problem in the clinical management of patients after transplantation. Immunosuppressants act on the cardiovascular system in a variety of ways, including fibrosis and myocardial remodeling, endothelium disfunction, hypertension, atherosclerosis, dyslipidemia or hyperglycaemia, metabolic syndrome, and hyperuricemia. The use of multidrug protocols makes it possible to develop regimens that can reduce the incidence of cardiovascular events. A better understanding of their mechanism of action and the range of complications could enable physicians to select the appropriate therapy for a given patient, as well as to reduce complications and prolong life.
Collapse
Affiliation(s)
- Bianka Opałka
- Department of Histology and Developmental Biology, Faculty of Health Sciences, Pomeranian Medical University, 71-210 Szczecin, Poland;
| | - Michał Żołnierczuk
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland;
| | - Marta Grabowska
- Department of Histology and Developmental Biology, Faculty of Health Sciences, Pomeranian Medical University, 71-210 Szczecin, Poland;
| |
Collapse
|
13
|
Stephenson EJ, Kinney CE, Stayton AS, Han JC. Energy expenditure deficits drive obesity in a mouse model of Alström syndrome. Obesity (Silver Spring) 2023; 31:2786-2798. [PMID: 37712194 DOI: 10.1002/oby.23877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 06/27/2023] [Indexed: 09/16/2023]
Abstract
OBJECTIVE Alström syndrome (AS) is a rare multisystem disorder of which early onset childhood obesity is a cardinal feature. Like humans with AS, animal models with Alms1 loss-of-function mutations develop obesity, supporting the notion that ALMS1 is required for the regulatory control of energy balance across species. This study aimed to determine which component(s) of energy balance are reliant on ALMS1. METHODS Comprehensive energy balance phenotyping was performed on Alms1tvrm102 mice at both 8 and 18 weeks of age. RESULTS It was found that adiposity gains occurred early and rapidly in Alms1tvrm102 male mice but much later in females. Rapid increases in body fat in males were due to a marked reduction in energy expenditure (EE) during early life and not due to any genotype-specific increases in energy intake under chow conditions. Energy intake did increase in a genotype-specific manner when mice were provided a high-fat diet, exacerbating the effects of reduced EE on obesity progression. The EE deficit observed in male Alms1tvrm102 mice did not persist as mice aged. CONCLUSIONS Either loss of ALMS1 causes a developmental delay in the mechanisms controlling early life EE or activation of compensatory mechanisms occurs after obesity is established in AS. Future studies will determine how ALMS1 modulates EE and how sex moderates this process.
Collapse
Affiliation(s)
- Erin J Stephenson
- Department of Anatomy, College of Graduate Studies and Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois, USA
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| | - Clint E Kinney
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| | - Amanda S Stayton
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee, USA
- Department of Surgery, College of Medicine, James D. Eason Transplant Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Joan C Han
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai and Kravis Children's Hospital, New York, New York, USA
| |
Collapse
|
14
|
Nedergaard J, von Essen G, Cannon B. Brown adipose tissue: can it keep us slim? A discussion of the evidence for and against the existence of diet-induced thermogenesis in mice and men. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220220. [PMID: 37661736 PMCID: PMC10475870 DOI: 10.1098/rstb.2022.0220] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/12/2023] [Indexed: 09/05/2023] Open
Abstract
The issue under discussion here is whether a decrease in the degree of UCP1 activity (and brown adipose tissue activity in general) could be a cause of obesity in humans. This possibility principally requires the existence of the phenomenon of diet-induced thermogenesis. Obesity could be a consequence of a reduced functionality of diet-induced thermogenesis. Experiments in mice indicate that diet-induced thermogenesis exists and is dependent on the presence of UCP1 and thus of brown adipose tissue activity. Accordingly, many (but not all) experiments indicate that in the absence of UCP1, mice become obese. Whether similar mechanisms exist in humans is still unknown. A series of studies have indicated a correlation between obesity and low brown adipose tissue activity, but it may be so that the obesity itself may influence the estimates of brown adipose tissue activity (generally glucose uptake), partly explaining the relationship. Estimates of brown adipose tissue catabolizing activity would seem to indicate that it may possess a capacity sufficient to help maintain body weight, and obesity would thus be aggravated in its absence. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part II)'.
Collapse
Affiliation(s)
- Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Gabriella von Essen
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| |
Collapse
|
15
|
Xu J, Shao X, Zeng H, Wang C, Li J, Peng X, Zhuo Y, Hua L, Meng F, Han X. Hepatic-Specific FGF21 Knockout Abrogates Ovariectomy-Induced Obesity by Reversing Corticosterone Production. Int J Mol Sci 2023; 24:14922. [PMID: 37834368 PMCID: PMC10573867 DOI: 10.3390/ijms241914922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Increased glucocorticoid (GC) levels act as a master contributor to central obesity in estrogen-depleted females; however, what factors cause their increased GC production is unclear. Given (1) liver fibroblast growth factor 21 (FGF21) and GCs regulate each other's production in a feed-forward loop, and (2) circulating FGF21 and GCs are parallelly increased in menopausal women and ovariectomized mice, we thus hypothesized that elevation of hepatic FGF21 secretion causes increased GGs production in estrogen-depleted females. Using the ovariectomized mice as a model for menopausal women, we found that ovariectomy (OVX) increased circulating corticosterone levels, which in turn increased visceral adipose Hsd11b1 expression, thus causing visceral obesity in females. In contrast, liver-specific FGF21 knockout (FGF21 LKO) completely reversed OVX-induced high GCs and high visceral adipose Hsd11b1 expression, thus abrogating OVX-induced obesity in females. Even though FGF21 LKO failed to rescue OVX-induced dyslipidemia, hepatic steatosis, and insulin resistance. What's worse, FGF21 LKO even further exacerbated whole-body glucose metabolic dysfunction as evidenced by more impaired glucose and pyruvate tolerance and worsened insulin resistance. Mechanically, we found that FGF21 LKO reduced circulating insulin levels, thus causing the dissociation between decreased central obesity and the improvement of obesity-related metabolic syndromes in OVX mice. Collectively, our results suggest that liver FGF21 plays an essential role in mediating OVX-induced central obesity by promoting GC production. However, lack of liver FGF21 signaling reduces insulin production and in turn causes the dissociation between decreased central obesity and the improvement of obesity-related metabolic syndromes, highlighting a detrimental role for hepatic FGF21 signals in mediating the development of central obesity but a beneficial role in preventing metabolic abnormality from further exacerbation in estrogen-depleted females.
Collapse
Affiliation(s)
- Jiayu Xu
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Xinyu Shao
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Haozhe Zeng
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Chengxi Wang
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Jiayi Li
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Xiaoqin Peng
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Yong Zhuo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611134, China (L.H.)
| | - Lun Hua
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611134, China (L.H.)
| | - Fengyan Meng
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Xingfa Han
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| |
Collapse
|
16
|
Maushart CI, Sun W, Othman A, Ghosh A, Senn JR, Fischer JGW, Madoerin P, Loeliger RC, Benz RM, Takes M, Zech CJ, Chirindel A, Beuschlein F, Reincke M, Wild D, Bieri O, Zamboni N, Wolfrum C, Betz MJ. Effect of high-dose glucocorticoid treatment on human brown adipose tissue activity: a randomised, double-blinded, placebo-controlled cross-over trial in healthy men. EBioMedicine 2023; 96:104771. [PMID: 37659283 PMCID: PMC10483510 DOI: 10.1016/j.ebiom.2023.104771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND Glucocorticoids (GCs) are widely applied anti-inflammatory drugs that are associated with adverse metabolic effects including insulin resistance and weight gain. Previous research indicates that GCs may negatively impact brown adipose tissue (BAT) activity in rodents and humans. METHODS We performed a randomised, double-blinded cross-over trial in 16 healthy men (clinicaltrials.govNCT03269747). Participants received 40 mg of prednisone per day for one week or placebo. After a washout period of four weeks, participants crossed-over to the other treatment arm. Primary endpoint was the increase in resting energy expenditure (EE) in response to a mild-cold stimulus (cold-induced thermogenesis, CIT). Secondary outcomes comprised mean 18F-FDG uptake into supraclavicular BAT (SUVmean) as determined by FDG-PET/CT, volume of the BAT depot as well as fat content determined by MRI. The plasma metabolome and the transcriptome of supraclavicular BAT and of skeletal muscle biopsies after each treatment period were analysed. FINDINGS Sixteen participants were recruited to the trial and completed it successfully per protocol. After prednisone treatment resting EE was higher both during warm and cold conditions. However, CIT was similar, 153 kcal/24 h (95% CI 40-266 kcal/24 h) after placebo and 186 kcal/24 h (95% CI 94-277 kcal/24 h, p = 0.38) after prednisone. SUVmean of BAT after cold exposure was not significantly affected by prednisone (3.36 g/ml, 95% CI 2.69-4.02 g/ml, vs 3.07 g/ml, 95% CI 2.52-3.62 g/ml, p = 0.28). Results of plasma metabolomics and BAT transcriptomics corroborated these findings. RNA sequencing of muscle biopsies revealed higher expression of genes involved in calcium cycling. No serious adverse events were reported and adverse events were evenly distributed between the two treatments. INTERPRETATION Prednisone increased EE in healthy men possibly by altering skeletal muscle calcium cycling. Cold-induced BAT activity was not affected by GC treatment, which indicates that the unfavourable metabolic effects of GCs are independent from thermogenic adipocytes. FUNDING Grants from Swiss National Science Foundation (PZ00P3_167823), Bangerter-Rhyner Foundation and from Nora van der Meeuwen-Häfliger Foundation to MJB. A fellowship-grant from the Swiss National Science Foundation (SNF211053) to WS. Grants from German Research Foundation (project number: 314061271-TRR 205) and Else Kröner-Fresenius (grant support 2012_A103 and 2015_A228) to MR.
Collapse
Affiliation(s)
- Claudia Irene Maushart
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland.
| | - Wenfei Sun
- Institute of Food, Nutrition, and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Alaa Othman
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
| | - Adhideb Ghosh
- Institute of Food, Nutrition, and Health, ETH Zurich, Schwerzenbach, Switzerland; Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland.
| | - Jaël Rut Senn
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland.
| | - Jonas Gabriel William Fischer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland.
| | - Philipp Madoerin
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Rahel Catherina Loeliger
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland.
| | - Robyn Melanie Benz
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland.
| | - Martin Takes
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Christoph Johannes Zech
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Alin Chirindel
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University Zurich (UZH), Zurich, Switzerland; Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany.
| | - Martin Reincke
- Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany.
| | - Damian Wild
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Oliver Bieri
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Nicola Zamboni
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
| | - Christian Wolfrum
- Institute of Food, Nutrition, and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Matthias Johannes Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland.
| |
Collapse
|
17
|
Jovanovic P, Pool AH, Morones N, Wang Y, Novinbakht E, Keshishian N, Jang K, Oka Y, Riera CE. A sex-specific thermogenic neurocircuit induced by predator smell recruiting cholecystokinin neurons in the dorsomedial hypothalamus. Nat Commun 2023; 14:4937. [PMID: 37582805 PMCID: PMC10427624 DOI: 10.1038/s41467-023-40484-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 07/31/2023] [Indexed: 08/17/2023] Open
Abstract
Olfactory cues are vital for prey animals like rodents to perceive and evade predators. Stress-induced hyperthermia, via brown adipose tissue (BAT) thermogenesis, boosts physical performance and facilitates escape. However, many aspects of this response, including thermogenic control and sex-specific effects, remain enigmatic. Our study unveils that the predator odor trimethylthiazoline (TMT) elicits BAT thermogenesis, suppresses feeding, and drives glucocorticoid release in female mice. Chemogenetic stimulation of olfactory bulb (OB) mitral cells recapitulates the thermogenic output of this response and associated stress hormone corticosterone release in female mice. Neuronal projections from OB to medial amygdala (MeA) and dorsomedial hypothalamus (DMH) exhibit female-specific cFos activity toward odors. Cell sorting and single-cell RNA-sequencing of DMH identify cholecystokinin (CCK)-expressing neurons as recipients of predator odor cues. Chemogenetic manipulation and neuronal silencing of DMHCCK neurons further implicate these neurons in the propagation of predator odor-associated thermogenesis and food intake suppression, highlighting their role in female stress-induced hyperthermia.
Collapse
Affiliation(s)
- Predrag Jovanovic
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Allan-Hermann Pool
- Department of Neuroscience, Department of Anesthesiology and Pain Management, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nancy Morones
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Yidan Wang
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Edward Novinbakht
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Nareg Keshishian
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Kaitlyn Jang
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Yuki Oka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Celine E Riera
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA.
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA.
- Department of Neurology, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA.
| |
Collapse
|
18
|
McCormack NM, Nguyen NY, Tully CB, Oliver T, Fiorillo AA, Heier CR. Vamorolone improves Becker muscular dystrophy and increases dystrophin protein in bmx model mice. iScience 2023; 26:107161. [PMID: 37534133 PMCID: PMC10391915 DOI: 10.1016/j.isci.2023.107161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/15/2023] [Accepted: 06/13/2023] [Indexed: 08/04/2023] Open
Abstract
There is no approved therapy for Becker muscular dystrophy (BMD), a genetic muscle disease caused by in-frame dystrophin deletions. We previously developed the dissociative corticosteroid vamorolone for treatment of the allelic, dystrophin-null disease Duchenne muscular dystrophy. We hypothesize vamorolone can treat BMD by safely reducing inflammatory signaling in muscle and through a novel mechanism of increasing dystrophin protein via suppression of dystrophin-targeting miRNAs. Here, we test this in the bmx mouse model of BMD. Daily oral treatment with vamorolone or prednisolone improves bmx grip strength and hang time phenotypes. Both drugs reduce myofiber size and decrease the percentage of centrally nucleated fibers. Vamorolone shows improved safety versus prednisolone by avoiding or reducing key side effects to behavior and growth. Intriguingly, vamorolone increases dystrophin protein in both heart and skeletal muscle. These data indicate that vamorolone, nearing approval for Duchenne, shows efficacy in bmx mice and therefore warrants clinical investigation in BMD.
Collapse
Affiliation(s)
- Nikki M. McCormack
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Nhu Y. Nguyen
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Christopher B. Tully
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Trinitee Oliver
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Biology, Howard University, Washington, DC, USA
| | - Alyson A. Fiorillo
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC, USA
| | - Christopher R. Heier
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC, USA
| |
Collapse
|
19
|
Wang S, Liu Y, Chen J, He Y, Ma W, Liu X, Sun X. Effects of multi-organ crosstalk on the physiology and pathology of adipose tissue. Front Endocrinol (Lausanne) 2023; 14:1198984. [PMID: 37383400 PMCID: PMC10293893 DOI: 10.3389/fendo.2023.1198984] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/26/2023] [Indexed: 06/30/2023] Open
Abstract
In previous studies, adipocytes were found to play an important role in regulating whole-body nutrition and energy balance, and are also important in energy metabolism, hormone secretion, and immune regulation. Different adipocytes have different contributions to the body, with white adipocytes primarily storing energy and brown adipocytes producing heat. Recently discovered beige adipocytes, which have characteristics in between white and brown adipocytes, also have the potential to produce heat. Adipocytes interact with other cells in the microenvironment to promote blood vessel growth and immune and neural network interactions. Adipose tissue plays an important role in obesity, metabolic syndrome, and type 2 diabetes. Dysfunction in adipose tissue endocrine and immune regulation can cause and promote the occurrence and development of related diseases. Adipose tissue can also secrete multiple cytokines, which can interact with organs; however, previous studies have not comprehensively summarized the interaction between adipose tissue and other organs. This article reviews the effect of multi-organ crosstalk on the physiology and pathology of adipose tissue, including interactions between the central nervous system, heart, liver, skeletal muscle, and intestines, as well as the mechanisms of adipose tissue in the development of various diseases and its role in disease treatment. It emphasizes the importance of a deeper understanding of these mechanisms for the prevention and treatment of related diseases. Determining these mechanisms has enormous potential for identifying new targets for treating diabetes, metabolic disorders, and cardiovascular diseases.
Collapse
Affiliation(s)
- Sufen Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yifan Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jiaqi Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yuejing He
- Clinical Laboratory, Dongguan Eighth People’s Hospital, Dongguan, China
| | - Wanrui Ma
- Department of General Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Xinguang Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Xuerong Sun
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| |
Collapse
|
20
|
Abstract
Brown adipose tissue (BAT) displays the unique capacity to generate heat through uncoupled oxidative phosphorylation that makes it a very attractive therapeutic target for cardiometabolic diseases. Here, we review BAT cellular metabolism, its regulation by the central nervous and endocrine systems and circulating metabolites, the plausible roles of this tissue in human thermoregulation, energy balance, and cardiometabolic disorders, and the current knowledge on its pharmacological stimulation in humans. The current definition and measurement of BAT in human studies relies almost exclusively on BAT glucose uptake from positron emission tomography with 18F-fluorodeoxiglucose, which can be dissociated from BAT thermogenic activity, as for example in insulin-resistant states. The most important energy substrate for BAT thermogenesis is its intracellular fatty acid content mobilized from sympathetic stimulation of intracellular triglyceride lipolysis. This lipolytic BAT response is intertwined with that of white adipose (WAT) and other metabolic tissues, and cannot be independently stimulated with the drugs tested thus far. BAT is an interesting and biologically plausible target that has yet to be fully and selectively activated to increase the body's thermogenic response and shift energy balance. The field of human BAT research is in need of methods able to directly, specifically, and reliably measure BAT thermogenic capacity while also tracking the related thermogenic responses in WAT and other tissues. Until this is achieved, uncertainty will remain about the role played by this fascinating tissue in human cardiometabolic diseases.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | | | - Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec City, Quebec, G1V 4G5, Canada
| |
Collapse
|
21
|
Chronic Corticosterone Exposure Suppresses Copper Transport through GR-Mediated Intestinal CTR1 Pathway in Mice. BIOLOGY 2023; 12:biology12020197. [PMID: 36829476 PMCID: PMC9953443 DOI: 10.3390/biology12020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023]
Abstract
Numerous studies have discovered that chronic stress induces metabolic disorders by affecting iron and zinc metabolism, but the relationship between chronic stress and copper metabolism remains unclear. Here, we explore the influence of chronic corticosterone (CORT) exposure on copper metabolism and its regulatory mechanism in mice. Mice were treated with 100 μg/mL CORT in drinking water for a 4-week trial. We found that CORT treatment resulted in a significant decrease in plasma copper level, plasma ceruloplasmin activity, plasma and liver Cu/Zn-SOD activity, hepatic copper content, and liver metallothionein content in mice. CORT treatment led to the reduction in duodenal expression of copper transporter 1 (CTR1), duodenal cytochrome b (DCYTB), and ATPase copper-transporting alpha (ATP7A) at the mRNA and protein level in mice. CORT treatment activated nuclear glucocorticoid receptor (GR) and down-regulated CRT1 expression in Caco-2 cells, whereas these phenotypes were reversible by an antagonist of GR, RU486. Chromatin immunoprecipitation analysis revealed that GR bound to the Ctr1 promoter in Caco-2 cells. Transient transfection assays in Caco-2 cells demonstrated that the Ctr1 promoter was responsive to the CORT-activated glucocorticoid receptor, whereas mutation/deletion of the glucocorticoid receptor element (GRE) markedly impaired activation of the Ctr1 promoter. In addition, CORT-induced downregulation of Ctr1 promoter activity was markedly attenuated in Caco-2 cells when RU486 was added. These findings present a novel molecular target for CORT that down-regulates intestinal CTR1 expression via GR-mediated trans-repression in mice.
Collapse
|
22
|
Zhou Z, Zhang A, Liu X, Yang Y, Zhao R, Jia Y. m 6A-Mediated PPARA Translational Suppression Contributes to Corticosterone-Induced Visceral Fat Deposition in Chickens. Int J Mol Sci 2022; 23:ijms232415761. [PMID: 36555401 PMCID: PMC9779672 DOI: 10.3390/ijms232415761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Excess fat deposition in broilers leads to great economic losses and is harmful to consumers' health. Chronic stress in the life cycle of chickens could be an important trigger. However, the underlying mechanisms are still unclear. In this study, 30-day-old chickens were subcutaneously injected with 2 mg/kg corticosterone (CORT) twice a day for 14 days to simulate long-term stress. It was shown that chronic CORT exposure significantly increased plasma triglyceride concentrations and enlarged the adipocyte sizes in chickens. Meanwhile, chronic CORT administration significantly enlarged the adipocyte sizes, increased the protein contents of FASN and decreased HSL, ATGL, Beclin1 and PPARA protein levels. Moreover, global m6A methylations were significantly reduced and accompanied by downregulated METTL3 and YTHDF2 protein expression by CORT treatment. Interestingly, the significant differences of site-specific m6A demethylation were observed in exon7 of PPARA mRNA. Additionally, a mutation of the m6A site in the PPARA gene fused GFP and revealed that demethylated RRACH in PPARA CDS impaired protein translation in vitro. In conclusion, these results indicated that m6A-mediated PPARA translational suppression contributes to CORT-induced visceral fat deposition in chickens, which may provide a new target for the treatment of Cushing's syndrome.
Collapse
Affiliation(s)
- Zixuan Zhou
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Aijia Zhang
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xinyi Liu
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yang Yang
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing 210095, China
| | - Yimin Jia
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing 210095, China
- Correspondence: ; Tel.: +86-2584396413; Fax: +86-2584398669
| |
Collapse
|
23
|
Tholen S, Patel R, Agas A, Kovary KM, Rabiee A, Nicholls HT, Bielczyk-Maczyńska E, Yang W, Kraemer FB, Teruel MN. Flattening of circadian glucocorticoid oscillations drives acute hyperinsulinemia and adipocyte hypertrophy. Cell Rep 2022; 39:111018. [PMID: 35767959 PMCID: PMC9391061 DOI: 10.1016/j.celrep.2022.111018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 04/14/2022] [Accepted: 06/08/2022] [Indexed: 11/03/2022] Open
Abstract
Disruption of circadian glucocorticoid oscillations in Cushing's disease and chronic stress results in obesity and adipocyte hypertrophy, which is believed to be a main source of the harmful effects of obesity. Here, we recapitulate stress due to jet lag or work-life imbalances by flattening glucocorticoid oscillations in mice. Within 3 days, mice achieve a metabolic state with persistently high insulin, but surprisingly low glucose and fatty acids in the bloodstream, that precedes a more than 2-fold increase in brown and white adipose tissue mass within 3 weeks. Transcriptomic and Cd36-knockout mouse analyses show that hyperinsulinemia-mediated de novo fatty acid synthesis and Cd36-mediated fatty acid uptake drive fat mass increases. Intriguingly, this mechanism by which glucocorticoid flattening causes acute hyperinsulinemia and adipocyte hypertrophy is unexpectedly beneficial in preventing high levels of circulating fatty acids and glucose for weeks, thus serving as a protective response to preserve metabolic health during chronic stress.
Collapse
Affiliation(s)
- Stefan Tholen
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Roma Patel
- Department of Biochemistry and the Gale and Ira Drukier Institute of Children's Health, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Agnieszka Agas
- Department of Biochemistry and the Gale and Ira Drukier Institute of Children's Health, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Kyle M Kovary
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Atefeh Rabiee
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Hayley T Nicholls
- Weill Center for Metabolic Health, Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Ewa Bielczyk-Maczyńska
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Wenting Yang
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Fredric B Kraemer
- Department of Medicine, Division of Endocrinology, Stanford University, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA 94305, USA
| | - Mary N Teruel
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Biochemistry and the Gale and Ira Drukier Institute of Children's Health, Weill Cornell Medical College of Cornell University, New York, NY, USA; Weill Center for Metabolic Health, Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA.
| |
Collapse
|
24
|
Bel JS, Tai TC, Khaper N, Lees SJ. Chronic glucocorticoid exposure causes brown adipose tissue whitening, alters whole-body glucose metabolism and increases tissue uncoupling protein-1. Physiol Rep 2022; 10:e15292. [PMID: 35510321 PMCID: PMC9069169 DOI: 10.14814/phy2.15292] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 04/14/2023] Open
Abstract
Adipose tissue (AT) has been found to exist in two predominant forms, white and brown. White adipose tissue (WAT) is the body's conventional storage organ, and brown adipose tissue (BAT) is responsible for non-shivering thermogenesis which allows mammals to produce heat and regulate body temperature. Studies examining BAT and its role in whole-body metabolism have found that active BAT utilizes glucose and circulating fatty acids and is associated with improved metabolic outcomes. While the beiging of WAT is a growing area of interest, the possibility of the BAT depot to "whiten" and store more triglycerides also has metabolic and health implications. Currently, there are limited studies that examine the effects of chronic stress and its ability to induce a white-like phenotype in the BAT depot. This research examined how chronic exposure to the murine stress hormone, corticosterone, for 4 weeks can affect the whitening process of BAT in C57BL/6 male mice. Separate treatments with mirabegron, a known β3-adrenergic receptor agonist, were used to directly compare the effects of corticosterone with a beiging phenotype. Corticosterone-treated mice had significantly higher body weight (p ≤ 0.05) and BAT mass (p ≤ 0.05), increased adipocyte area (p ≤ 0.05), were insulin resistant (p ≤ 0.05), and significantly elevated expressions of uncoupling protein 1 (UCP-1) in BAT (p ≤ 0.05) while mitochondrial content remained unchanged. This whitened phenotype has not been previously associated with increased uncoupling proteins under chronic stress and may represent a compensatory mechanism being initiated under these conditions. These findings have implications for the study of BAT in response to chronic glucocorticoid exposure potentially leading to BAT dysfunction and negative impacts on whole-body glucose metabolism.
Collapse
Affiliation(s)
- Jocelyn S. Bel
- Biotechnology ProgramLakehead UniversityThunder BayOntarioCanada
| | - T. C. Tai
- Northern Ontario School of MedicineThunder BayOntarioCanada
- BiologyLaurentian UniversitySudburyOntarioCanada
- Chemistry and BiochemistryLaurentian UniversitySudburyOntarioCanada
- Biomolecular Sciences ProgramLaurentian UniversitySudburyOntarioCanada
| | - Neelam Khaper
- Northern Ontario School of MedicineThunder BayOntarioCanada
- Biomolecular Sciences ProgramLaurentian UniversitySudburyOntarioCanada
- BiologyLakehead UniversityThunder BayOntarioCanada
| | - Simon J. Lees
- Northern Ontario School of MedicineThunder BayOntarioCanada
- BiologyLakehead UniversityThunder BayOntarioCanada
| |
Collapse
|
25
|
Countering obesity with eosinophils and sympathetic fat. Proc Natl Acad Sci U S A 2022; 119:2123084119. [PMID: 35105817 PMCID: PMC8833142 DOI: 10.1073/pnas.2123084119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
26
|
Abstract
Concerning diet-induced thermogenesis, methodological issues relate mainly to the interpretation of measurements, rather than to the technical methodology as such. In the following, we point to a series of issues where the analysis often suggests the occurrence of UCP1-related diet-induced thermogenesis but where the observations are often the consequences of a process that has induced leanness rather than being the cause of them. We particularly emphasize the necessity of focusing on the total organism when interpreting biochemical and molecular data, where the concept of total tissue values rather than relative data better reflects physiologically important alterations. We stress the importance of performing experiments at thermoneutrality in order to obtain clinically relevant data and stress that true thermogenic agents may be overlooked if this is not done.
Collapse
Affiliation(s)
- Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
27
|
Shamsi F, Wang CH, Tseng YH. The evolving view of thermogenic adipocytes - ontogeny, niche and function. Nat Rev Endocrinol 2021; 17:726-744. [PMID: 34625737 PMCID: PMC8814904 DOI: 10.1038/s41574-021-00562-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 12/12/2022]
Abstract
The worldwide incidence of obesity and its sequelae, such as type 2 diabetes mellitus, have reached pandemic levels. Central to the development of these metabolic disorders is adipose tissue. White adipose tissue stores excess energy, whereas brown adipose tissue (BAT) and beige (also known as brite) adipose tissue dissipate energy to generate heat in a process known as thermogenesis. Strategies that activate and expand BAT and beige adipose tissue increase energy expenditure in animal models and offer therapeutic promise to treat obesity. A better understanding of the molecular mechanisms underlying the development of BAT and beige adipose tissue and the activation of thermogenic function is the key to creating practical therapeutic interventions for obesity and metabolic disorders. In this Review, we discuss the regulation of the tissue microenvironment (the adipose niche) and inter-organ communication between BAT and other tissues. We also cover the activation of BAT and beige adipose tissue in response to physiological cues (such as cold exposure, exercise and diet). We highlight advances in harnessing the therapeutic potential of BAT and beige adipose tissue by genetic, pharmacological and cell-based approaches in obesity and metabolic disorders.
Collapse
Affiliation(s)
- Farnaz Shamsi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA
| | - Chih-Hao Wang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
28
|
Luo L, Wang L, Luo Y, Romero E, Yang X, Liu M. Glucocorticoid/Adiponectin Axis Mediates Full Activation of Cold-Induced Beige Fat Thermogenesis. Biomolecules 2021; 11:1573. [PMID: 34827571 PMCID: PMC8615797 DOI: 10.3390/biom11111573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoids (GCs), a class of corticosteroids produced by the adrenal cortex in response to stress, exert obesity-promoting effects. Although adaptive thermogenesis has been considered an effective approach to counteract obesity, whether GCs play a role in regulating cold stress-induced thermogenesis remains incompletely understood. Here, we show that the circulating levels of stress hormone corticosterone (GC in rodents) were significantly elevated, whereas the levels of adiponectin, an adipokine that was linked to cold-induced adaptive thermogenesis, were decreased 48 h post cold exposure. The administration of a glucocorticoid hydrocortisone downregulated adiponectin protein and mRNA levels in both WAT and white adipocytes, and upregulated thermogenic gene expression in inguinal fat. In contrast, mifepristone, a glucocorticoid receptor antagonist, enhanced adiponectin expression and suppressed energy expenditure in vivo. Mechanistically, hydrocortisone suppressed adiponectin expression by antagonizing PPARγ in differentiated 3T3-L1 adipocytes. Ultimately, adiponectin deficiency restored mifepristone-decreased oxygen consumption and suppressed the expression of thermogenic genes in inguinal fat. Taken together, our study reveals that the GCs/adiponectin axis is a key regulator of beige fat thermogenesis in response to acute cold stress.
Collapse
Affiliation(s)
- Liping Luo
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (L.L.); (L.W.); (Y.L.); (E.R.); (X.Y.)
| | - Lu Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (L.L.); (L.W.); (Y.L.); (E.R.); (X.Y.)
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yan Luo
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (L.L.); (L.W.); (Y.L.); (E.R.); (X.Y.)
- Department of Endocrinology and Metabolism, Metabolic Syndrome Research Center, The Second Xiangya Hospital, Central South University, Changsha 410011, China
- Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Estevan Romero
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (L.L.); (L.W.); (Y.L.); (E.R.); (X.Y.)
| | - Xin Yang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (L.L.); (L.W.); (Y.L.); (E.R.); (X.Y.)
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (L.L.); (L.W.); (Y.L.); (E.R.); (X.Y.)
- Autophagy, Inflammation and Metabolism Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|
29
|
Genomic and Non-Genomic Actions of Glucocorticoids on Adipose Tissue Lipid Metabolism. Int J Mol Sci 2021; 22:ijms22168503. [PMID: 34445209 PMCID: PMC8395154 DOI: 10.3390/ijms22168503] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/01/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Glucocorticoids (GCs) are hormones that aid the body under stress by regulating glucose and free fatty acids. GCs maintain energy homeostasis in multiple tissues, including those in the liver and skeletal muscle, white adipose tissue (WAT), and brown adipose tissue (BAT). WAT stores energy as triglycerides, while BAT uses fatty acids for heat generation. The multiple genomic and non-genomic pathways in GC signaling vary with exposure duration, location (adipose tissue depot), and species. Genomic effects occur directly through the cytosolic GC receptor (GR), regulating the expression of proteins related to lipid metabolism, such as ATGL and HSL. Non-genomic effects act through mechanisms often independent of the cytosolic GR and happen shortly after GC exposure. Studying the effects of GCs on adipose tissue breakdown and generation (lipolysis and adipogenesis) leads to insights for treatment of adipose-related diseases, such as obesity, coronary disease, and cancer, but has led to controversy among researchers, largely due to the complexity of the process. This paper reviews the recent literature on the genomic and non-genomic effects of GCs on WAT and BAT lipolysis and proposes research to address the many gaps in knowledge related to GC activity and its effects on disease.
Collapse
|
30
|
Morris EM, Noland RD, Ponte ME, Montonye ML, Christianson JA, Stanford JA, Miles JM, Hayes MR, Thyfault JP. Reduced Liver-Specific PGC1a Increases Susceptibility for Short-Term Diet-Induced Weight Gain in Male Mice. Nutrients 2021; 13:2596. [PMID: 34444756 PMCID: PMC8400659 DOI: 10.3390/nu13082596] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 11/18/2022] Open
Abstract
The central integration of peripheral neural signals is one mechanism by which systemic energy homeostasis is regulated. Previously, increased acute food intake following the chemical reduction of hepatic fatty acid oxidation and ATP levels was prevented by common hepatic branch vagotomy (HBV). However, possible offsite actions of the chemical compounds confound the precise role of liver energy metabolism. Herein, we used a hepatocyte PGC1a heterozygous (LPGC1a) mouse model, with associated reductions in mitochondrial fatty acid oxidation and respiratory capacity, to assess the role of liver energy metabolism in systemic energy homeostasis. LPGC1a male, but not female, mice had a 70% greater high-fat/high-sucrose (HFHS) diet-induced weight gain compared to wildtype (WT) mice (p < 0.05). The greater weight gain was associated with altered feeding behavior and lower activity energy expenditure during the HFHS diet in LPGC1a males. WT and LPGC1a mice underwent sham surgery or HBV to assess whether vagal signaling was involved in the HFHS-induced weight gain of male LPGC1a mice. HBV increased HFHS-induced weight gain (85%, p < 0.05) in male WT mice, but not LPGC1a mice. These data demonstrate a sex-specific role of reduced liver energy metabolism in acute diet-induced weight gain, and the need for a more nuanced assessment of the role of vagal signaling in short-term diet-induced weight gain.
Collapse
Affiliation(s)
- E. Matthew Morris
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.D.N.); (M.E.P.); (J.A.S.); (J.P.T.)
- Center for Children’s Healthy Lifestyle and Nutrition, Children’s Mercy Hospital, Kansas City, MO 64108, USA
| | - Roberto D. Noland
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.D.N.); (M.E.P.); (J.A.S.); (J.P.T.)
| | - Michael E. Ponte
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.D.N.); (M.E.P.); (J.A.S.); (J.P.T.)
| | - Michelle L. Montonye
- Department of Nutrition & Exercise Physiology, University of Missouri, Columbia, MO 65211, USA;
| | - Julie A. Christianson
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - John A. Stanford
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.D.N.); (M.E.P.); (J.A.S.); (J.P.T.)
| | - John M. Miles
- Department of Internal Medicine—Division of Endocrinology and Metabolism, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Matthew R. Hayes
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - John P. Thyfault
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.D.N.); (M.E.P.); (J.A.S.); (J.P.T.)
- Center for Children’s Healthy Lifestyle and Nutrition, Children’s Mercy Hospital, Kansas City, MO 64108, USA
- Department of Internal Medicine—Division of Endocrinology and Metabolism, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Kansas City VA Medical Center-Research Service, Kansas City, MO 64128, USA
| |
Collapse
|
31
|
Neuroendocrine control of appetite and metabolism. Exp Mol Med 2021; 53:505-516. [PMID: 33837263 PMCID: PMC8102538 DOI: 10.1038/s12276-021-00597-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/11/2021] [Accepted: 02/17/2021] [Indexed: 02/02/2023] Open
Abstract
Body homeostasis is predominantly controlled by hormones secreted by endocrine organs. The central nervous system contains several important endocrine structures, including the hypothalamic-pituitary axis. Conventionally, neurohormones released by the hypothalamus and the pituitary gland (hypophysis) have received much attention owing to the unique functions of the end hormones released by their target peripheral organs (e.g., glucocorticoids released by the adrenal glands). Recent advances in mouse genetics have revealed several important metabolic functions of hypothalamic neurohormone-expressing cells, many of which are not readily explained by the action of the corresponding classical downstream hormones. Notably, the newly identified functions are better explained by the action of conventional neurotransmitters (e.g., glutamate and GABA) that constitute a neuronal circuit. In this review, we discuss the regulation of appetite and metabolism by hypothalamic neurohormone-expressing cells, with a focus on the distinct contributions of neurohormones and neurotransmitters released by these neurons.
Collapse
|
32
|
van Beek SMM, Kalinovich A, Schaart G, Bengtsson T, Hoeks J. Prolonged β 2-adrenergic agonist treatment improves glucose homeostasis in diet-induced obese UCP1 -/- mice. Am J Physiol Endocrinol Metab 2021; 320:E619-E628. [PMID: 33522400 DOI: 10.1152/ajpendo.00324.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Prolonged supplementation with the β2-agonist clenbuterol improves glucose homeostasis in diabetic rodents, likely via β2-adrenoceptor (β2-AR)-mediated effects in the skeletal muscle and liver. However, since rodents have, in contrast to-especially diabetic-humans, substantial quantities of brown adipose tissue (BAT) and clenbuterol has affinity to β1- and β3-ARs, the contribution of BAT to these improvements is unclear. Therefore, we investigated clenbuterol-mediated improvements in glucose homeostasis in uncoupling protein 1-deficient (UCP1-/-) mice, lacking thermogenic BAT, versus wild-type (WT) mice. Anesthetized WT and UCP1-/- C57Bl/6 mice were injected with saline or clenbuterol and whole body oxygen consumption was measured. Furthermore, male WT and UCP1-/- C57Bl/6 mice were subjected to 17-wk of chow feeding, high-fat feeding, or high-fat feeding with clenbuterol treatment between weeks 13 and 17. Body composition was measured weekly with MRI. Oral glucose tolerance and insulin tolerance tests were performed in week 15 and 17, respectively. Clenbuterol increased oxygen consumption approximately twofold in WT mice. This increase was blunted in UCP1-/- mice, indicating clenbuterol-mediated activation of BAT thermogenesis. High-fat feeding induced diabetogenic phenotypes in both genotypes. However, low-dose clenbuterol treatment for 2 wk significantly reduced fasting blood glucose by 12.9% in WT and 14.8% in UCP1-/- mice. Clenbuterol treatment improved glucose and insulin tolerance in both genotypes compared with HFD controls and normalized to chow-fed control mice independent of body mass and composition alterations. Clenbuterol improved whole body glucose homeostasis independent of UCP1. Given the low human abundancy of BAT, β2-AR agonist treatment provides a potential novel route for glucose disposal in diabetic humans.NEW & NOTEWORTHY Improvements in whole body glucose homeostasis of rodents upon prolonged β2-adrenergic agonist supplementation could potentially be attributed to UCP1-mediated BAT thermogenesis. Indeed, we show that acute injection with the β2-AR agonist clenbuterol induces BAT activation in mice. However, we also demonstrate that prolonged clenbuterol supplementation robustly improves whole body glucose and insulin tolerance in a similar way in both DIO WT and UCP1-/- mice, indicating that β2-AR agonist supplementation improves whole body glucose homeostasis independent of UCP1-mediated BAT thermogenesis.
Collapse
Affiliation(s)
- Sten M M van Beek
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Anastasia Kalinovich
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Gert Schaart
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Joris Hoeks
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
33
|
Felder M, Maushart CI, Gashi G, Senn JR, Becker AS, Müller J, Balaz M, Wolfrum C, Burger IA, Betz MJ. Fluvastatin Reduces Glucose Tolerance in Healthy Young Individuals Independently of Cold Induced BAT Activity. Front Endocrinol (Lausanne) 2021; 12:765807. [PMID: 34858338 PMCID: PMC8631514 DOI: 10.3389/fendo.2021.765807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/14/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Statins are commonly prescribed for primary and secondary prevention of atherosclerotic disease. They reduce cholesterol biosynthesis by inhibiting hydroxymethylglutaryl-coenzyme A-reductase (HMG-CoA-reductase) and therefore mevalonate synthesis. Several studies reported a small, but significant increase in the diagnosis of diabetes mellitus with statin treatment. The molecular mechanisms behind this adverse effect are not yet fully understood. Brown adipose tissue (BAT), which plays a role in thermogenesis, has been associated with a reduced risk of insulin resistance. Statins inhibit adipose tissue browning and have been negatively linked to the presence of BAT in humans. We therefore speculated that inhibition of BAT by statins contributes to increased insulin resistance in humans. METHODS A prospective study was conducted in 17 young, healthy men. After screening whether significant cold-induced thermogenesis (CIT) was present, participants underwent glucose tolerance testing (oGTT) and assessment of BAT activity by FDG-PET/MRI after cold-exposure and treatment with a β3-agonist. Fluvastatin 2x40mg per day was then administered for two weeks and oGTT and FDG-PET/MRI were repeated. RESULTS Two weeks of fluvastatin treatment led to a significant increase in glucose area under the curve (AUC) during oGTT (p=0.02), reduction in total cholesterol and LDL cholesterol (both p<0.0001). Insulin AUC (p=0.26), resting energy expenditure (REE) (p=0.44) and diet induced thermogenesis (DIT) (p=0.27) did not change significantly. The Matsuda index, as an indicator of insulin sensitivity, was lower after fluvastatin intake, but the difference was not statistically significant (p=0.09). As parameters of BAT activity, mean standard uptake value (SUVmean) (p=0.12), volume (p=0.49) and total glycolysis (p=0.74) did not change significantly during the intervention. Matsuda index, was inversely related to SUVmean and the respiratory exchange ratio (RER) (both R2 = 0.44, p=0.005) at baseline, but not after administration of fluvastatin (R2 = 0.08, p=0.29, and R2 = 0.14, p=0.16, respectively). CONCLUSIONS Treatment with fluvastatin for two weeks reduced serum lipid levels but increased glucose AUC in young, healthy men, indicating reduced glucose tolerance. This was not associated with changes in cold-induced BAT activity.
Collapse
Affiliation(s)
- Martina Felder
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Claudia Irene Maushart
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Gani Gashi
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jaël Rut Senn
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Anton S. Becker
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich/University of Zurich, Zurich, Switzerland
| | - Julian Müller
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich/University of Zurich, Zurich, Switzerland
| | - Miroslav Balaz
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Irene A. Burger
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich/University of Zurich, Zurich, Switzerland
| | - Matthias Johannes Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
- *Correspondence: Matthias Johannes Betz,
| |
Collapse
|
34
|
Kaikaew K, Grefhorst A, Visser JA. Sex Differences in Brown Adipose Tissue Function: Sex Hormones, Glucocorticoids, and Their Crosstalk. Front Endocrinol (Lausanne) 2021; 12:652444. [PMID: 33927694 PMCID: PMC8078866 DOI: 10.3389/fendo.2021.652444] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022] Open
Abstract
Excessive fat accumulation in the body causes overweight and obesity. To date, research has confirmed that there are two types of adipose tissue with opposing functions: lipid-storing white adipose tissue (WAT) and lipid-burning brown adipose tissue (BAT). After the rediscovery of the presence of metabolically active BAT in adults, BAT has received increasing attention especially since activation of BAT is considered a promising way to combat obesity and associated comorbidities. It has become clear that energy homeostasis differs between the sexes, which has a significant impact on the development of pathological conditions such as type 2 diabetes. Sex differences in BAT activity may contribute to this and, therefore, it is important to address the underlying mechanisms that contribute to sex differences in BAT activity. In this review, we discuss the role of sex hormones in the regulation of BAT activity under physiological and some pathological conditions. Given the increasing number of studies suggesting a crosstalk between sex hormones and the hypothalamic-pituitary-adrenal axis in metabolism, we also discuss this crosstalk in relation to sex differences in BAT activity.
Collapse
Affiliation(s)
- Kasiphak Kaikaew
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Aldo Grefhorst
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
| | - Jenny A. Visser
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- *Correspondence: Jenny A. Visser,
| |
Collapse
|
35
|
Chitraju C, Fischer AW, Farese RV, Walther TC. Lipid Droplets in Brown Adipose Tissue Are Dispensable for Cold-Induced Thermogenesis. Cell Rep 2020; 33:108348. [PMID: 33147469 PMCID: PMC7696656 DOI: 10.1016/j.celrep.2020.108348] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/29/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022] Open
Abstract
Brown adipocytes store metabolic energy as triglycerides (TGs) in lipid droplets (LDs). Fatty acids released from brown adipocyte LDs by lipolysis are thought to activate and fuel UCP1-mediated thermogenesis. Here, we test this hypothesis by preventing fatty acid storage in murine brown adipocytes through brown adipose tissue (BAT)-specific deletions of the TG synthesis enzymes DGAT1 and DGAT2 (BA-DGAT KO). Despite the absence of TGs in brown adipocytes, BAT is functional, and BA-DGAT-KO mice maintain euthermia during acute or chronic cold exposure. As apparent adaptations to the lack of TG, brown adipocytes of BA-DGAT-KO mice appear to use circulating glucose and fatty acids, and stored glycogen, to fuel thermogenesis. Moreover, BA-DGAT-KO mice are resistant to diet-induced glucose intolerance, likely because of increased glucose disposal by BAT. We conclude that TGs in BAT are dispensable for its contribution to cold-induced thermogenesis, at least when other fuel sources are available.
Collapse
Affiliation(s)
- Chandramohan Chitraju
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Alexander W Fischer
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Robert V Farese
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Tobias C Walther
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA.
| |
Collapse
|
36
|
Kroon J, Pereira AM, Meijer OC. Glucocorticoid Sexual Dimorphism in Metabolism: Dissecting the Role of Sex Hormones. Trends Endocrinol Metab 2020; 31:357-367. [PMID: 32037025 DOI: 10.1016/j.tem.2020.01.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/20/2019] [Accepted: 01/13/2020] [Indexed: 12/15/2022]
Abstract
Glucocorticoids are steroid hormones that are of pivotal importance in human physiology. Glucocorticoid signaling is complex in nature and dependent on many interacting factors. As glucocorticoids exhibit sexually dimorphic effects on several key processes including in metabolism, crosstalk with the sex steroid hormones (androgens and estrogens) is relevant. In this review, we highlight the state-of-the-art knowledge on glucocorticoid sexual dimorphism and sex hormone crosstalk. We include current insight in the molecular mechanisms that underlie nuclear steroid receptor crosstalk, and sex hormone effects on glucocorticoid metabolism. Finally, we show how these findings translate to humans exposed to excess glucocorticoid signaling, and we propose future avenues in the emerging field of steroid hormone crosstalk.
Collapse
Affiliation(s)
- Jan Kroon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | - Alberto M Pereira
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
37
|
Xie X, Xiao Q, Xiong Z, Yu C, Zhou J, Fu Z. Crocin-I ameliorates the disruption of lipid metabolism and dysbiosis of the gut microbiota induced by chronic corticosterone in mice. Food Funct 2020; 10:6779-6791. [PMID: 31576875 DOI: 10.1039/c9fo01533g] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glucocorticoids (GCs) are widely used as anti-inflammatory and immunosuppressive drugs. However, chronic treatment with GCs in clinical settings has a series of side effects, such as metabolic disorders, gut microbiota dysbiosis and neurological impairment. Therefore, searching for a functional substance that can alleviate these side effects is greatly meaningful to clinical patients. Crocin is the main active ingredient of saffron, which has been reported to have numerous pharmacological activities. However, the action of crocin-I, one major member of the crocin family, on the physiological mediation in the individuals receiving GC treatment remains unclear. In this study, we aimed to evaluate the efficacy of crocin-I on lipid metabolism and the gut microbiota in a mouse model of chronic corticosterone (CORT) treatment. Our findings showed that crocin-I reduced the levels of triglycerides and total cholesterol and the ratio of low density lipoprotein to high density lipoprotein in the serum of CORT-treated mice. In addition, transcriptome analysis revealed that crocin-I was effective in mediating the amelioration of lipid metabolism, mainly in fatty acid metabolism and steroid biosynthesis in CORT-treated mice. Moreover, metabolome analysis demonstrated that crocin-I could restore the disturbed metabolites in the liver of CORT-treated mice, most of which are long-chain fatty acids. Furthermore, high-throughput sequencing of 16s rRNA revealed that crocin-I could mitigate the dysbiosis of the gut microbiota caused by CORT at a dose of 40 mg kg-1, by resulting in a significant increase in the alpha diversity of the microbes in the cecal contents and a significant reduction in the abundance of Firmicutes, whereas by increasing the abundance of Bacteroidetes. These results indicated that oral administration of crocin-I could modify the composition of the gut microbiota and alleviate hepatic lipid disorder in mice treated with a high dose of GCs.
Collapse
Affiliation(s)
- Xiaoxian Xie
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| | | | | | | | | | | |
Collapse
|
38
|
Nicolas S, Rochet N, Gautier N, Chabry J, Pisani DF. The adiponectin receptor agonist AdipoRon normalizes glucose metabolism and prevents obesity but not growth retardation induced by glucocorticoids in young mice. Metabolism 2020; 103:154027. [PMID: 31778708 DOI: 10.1016/j.metabol.2019.154027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/08/2019] [Accepted: 11/22/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Glucocorticoids (GCs) are highly effective anti-inflammatory and immunosuppressive drugs. However, prolonged GC therapy may cause numerous adverse effects leading to diabetes and obesity, as well as bone disorders such as osteoporosis in adults and growth retardation in children and adolescents. Prevention and care of the GC-induced adverse effects remain challenging. We have previously demonstrated the efficacy of a treatment with a non-peptidic agonist of adiponectin receptors, AdipoRon, to reverse behaviour disorders and fat mass gain induced by long-term GC treatment. In this work, we have established a relevant model of GC-induced growth and metabolic disorders and determined that AdipoRon is a potential therapeutic tool to reverse these metabolic disturbances. METHODS 5-Week-old mice were treated continuously with or without corticosterone (35 mg/L) in drinking water for seven consecutive weeks. Taking advantage of this mouse model displaying various growth and metabolic disorders, we assayed whether AdipoRon (daily intraperitoneal injection of 1 mg/kg/day for the last 20 days) might prevent the GC-induced adverse effects. The control group was treated with vehicle only. Nutritional behaviors and metabolic parameters were followed-up throughout the treatment. Serum insulin and leptin levels were measured by ELISA. Computed tomography and histological analysis of adipose tissue were assessed at the end of the experimental procedure. RESULTS We found that GC treatment in young mice resulted in continuously increased body weight gain associated with a food intake increase. Compared to vehicle-, GC-treated mice displayed early major hyperleptinemia (up to 6-fold more) and hyperinsulinemia (up to 20-fold more) maintained throughout the treatment. At the end of the experimental procedure, GC-treated mice displayed bone growth retardation (e.g. femur length 15.1 versus 14.0 mm, P < 0.01), higher abdominal adipose tissue volume (4.1 versus 2.3, P < 0.01) and altered glucose metabolism compared to control mice. Interestingly, AdipoRon prevented GC-induced effects on energy metabolism such as abdominal adiposity, insulinemia and leptinemia. However, AdipoRon failed to counteract bone growth retardation. CONCLUSION We characterized the very early pathological steps induced by long-term GC in young mice in a relevant model, including growth retardation, fat mass gain and glucose homeostasis dysregulation. The adiponectin system stimulation enabled normalization of the adipose tissue and metabolic features of GC-treated mice. Adiponectin receptor agonists such as AdipoRon might constitute a novel way to counteract some GC-induced adverse effects.
Collapse
Affiliation(s)
- Sarah Nicolas
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire UMR 7275, 660 route des lucioles, Sophia Antipolis, 06560 Valbonne, France
| | - Nathalie Rochet
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose UMR 7277, 28 avenue de Valombrose, 06107 Nice, France
| | - Nadine Gautier
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose UMR 7277, 28 avenue de Valombrose, 06107 Nice, France
| | - Joëlle Chabry
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire UMR 7275, 660 route des lucioles, Sophia Antipolis, 06560 Valbonne, France.
| | - Didier F Pisani
- Université Côte d'Azur, CNRS, Laboratoire de PhysioMédecine Moléculaire UMR7370, 28 avenue de Valombrose, 06107 Nice, France.
| |
Collapse
|
39
|
Valavi E, Javaherizadeh H, Hakimzadeh M, Amoori P. Improvement Of Congenital Chloride Diarrhea With Corticosteroids: An Incidental Finding. Pediatric Health Med Ther 2019; 10:153-156. [PMID: 31827341 PMCID: PMC6901380 DOI: 10.2147/phmt.s220725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/08/2019] [Indexed: 11/23/2022] Open
Abstract
Congenital chloride diarrhea of infancy is a life threatening disease. We discuss two boys with congenital chloride diarrhea over a long time period before and after kidney transplantation. In the first case, prenatal sonography revealed polyhydramnios and generalized bowel loop distention. The genetic study confirmed congenital chloride diarrhea of infancy. Multiple episodes of severe dehydration, hyponatremia and acute tubular necrosis were seen during the follow up period. He underwent a year of hemodialysis before kidney transplantation. Three periods of improvement concerning diarrhea occurred with the use of corticosteroids, taken for other reasons. These improvements were seen after prednisolone administration for mastoiditis and following prednisolone administration for kidney transplantation. The second case was a 3.5 year old boy who is the cousin of the first case. He was referred to hospital with chronic watery diarrhea, metabolic alkalosis, hypokalemia, hyponatremia and failure to thrive in the first year of life. He was also treated with prednisolone and showed significant improvement.
Collapse
Affiliation(s)
- Ehsan Valavi
- Department of Pediatric Nephrology, Abuzar Children’s Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hazhir Javaherizadeh
- Alimentary Tract Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehran Hakimzadeh
- Department of Pediatric Gastroenterology, Abuzar Children’s Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Parisa Amoori
- Department of Pediatric Nephrology, Abuzar Children’s Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
40
|
Townsend LK, Weber AJ, Barbeau PA, Holloway GP, Wright DC. Reactive oxygen species-dependent regulation of pyruvate dehydrogenase kinase-4 in white adipose tissue. Am J Physiol Cell Physiol 2019; 318:C137-C149. [PMID: 31721616 DOI: 10.1152/ajpcell.00313.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are important signaling molecules mediating the exercise-induced adaptations in skeletal muscle. Acute exercise also drives the expression of genes involved in reesterification and glyceroneogenesis in white adipose tissue (WAT), but whether ROS play any role in this effect has not been explored. We speculated that exercise-induced ROS would regulate acute exercise-induced responses in WAT. To address this question, we utilized various models to alter redox signaling in WAT. We examined basal and exercise-induced gene expression in a genetically modified mouse model of reduced mitochondrial ROS emission [mitochondrial catalase overexpression (MCAT)]. Additionally, H2O2, various antioxidants, and the β3-adrenergic receptor agonist CL316243 were used to assess gene expression in white adipose tissue culture. MCAT mice have reduced ROS emission from WAT, enlarged WAT depots and adipocytes, and greater pyruvate dehydrogenase kinase-4 (Pdk4) gene expression. In WAT culture, H2O2 reduced glyceroneogenic gene expression. In wild-type mice, acute exercise induced dramatic but transient increases in Pdk4 and phosphoenolpyruvate carboxykinase (Pck1) mRNA in both subcutaneous inguinal WAT and epididymal WAT depots, which was almost completely absent in MCAT mice. Furthermore, the induction of Pdk4 and Pck1 in WAT culture by CL316243 was markedly reduced in the presence of antioxidants N-acetyl-cysteine or vitamin E. Genetic and nutritional approaches that attenuate redox signaling prevent exercise- and β-agonist-induced gene expression within WAT. Combined, these data suggest that ROS represent important mediators of gene expression within WAT.
Collapse
Affiliation(s)
- Logan K Townsend
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Alyssa J Weber
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Pierre-Andre Barbeau
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Graham P Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - David C Wright
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
41
|
Glantschnig C, Mattijssen F, Vogl ES, Ali Khan A, Rios Garcia M, Fischer K, Müller T, Uhlenhaut H, Nawroth P, Scheideler M, Rose AJ, Pellegata N, Herzig S. The glucocorticoid receptor in brown adipocytes is dispensable for control of energy homeostasis. EMBO Rep 2019; 20:e48552. [PMID: 31559673 PMCID: PMC6832000 DOI: 10.15252/embr.201948552] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/02/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
Aberrant activity of the glucocorticoid (GC)/glucocorticoid receptor (GR) endocrine system has been linked to obesity-related metabolic dysfunction. Traditionally, the GC/GR axis has been believed to play a crucial role in adipose tissue formation and function in both, white (WAT) and brown adipose tissue (BAT). While recent studies have challenged this notion for WAT, the contribution of GC/GR signaling to BAT-dependent energy homeostasis remained unknown. Here, we have generated and characterized a BAT-specific GR-knockout mouse (GRBATKO ), for the first time allowing to genetically interrogate the metabolic impact of BAT-GR. The HPA axis in GRBATKO mice was intact, as was the ability of mice to adapt to cold. BAT-GR was dispensable for the adaptation to fasting-feeding cycles and the development of diet-induced obesity. In obesity, glucose and lipid metabolism, insulin sensitivity, and food intake remained unchanged, aligning with the absence of changes in thermogenic gene expression. Together, we demonstrate that the GR in UCP1-positive BAT adipocytes plays a negligible role in systemic metabolism and BAT function, thereby opposing a long-standing paradigm in the field.
Collapse
|
42
|
Luijten IHN, Cannon B, Nedergaard J. Glucocorticoids and Brown Adipose Tissue: Do glucocorticoids really inhibit thermogenesis? Mol Aspects Med 2019; 68:42-59. [PMID: 31323252 DOI: 10.1016/j.mam.2019.07.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022]
Abstract
A reduction in the thermogenic activity of brown adipose tissue (BAT) is presently discussed as a possible determinant for the development of obesity in humans. One group of endogenous factors that could potentially affect BAT activity is the glucocorticoids (e.g. cortisol). We analyse here studies examining the effects of alterations in glucocorticoid signaling on BAT recruitment and thermogenic capacity. We find that irrespective of which manipulation of glucocorticoid signaling is examined, a seemingly homogeneous picture of lowered thermogenic capacity due to glucocorticoid stimulation is apparently obtained: e.g. lowered uncoupling protein 1 (UCP1) protein levels per mg protein, and an increased lipid accumulation in BAT. However, further analyses generally indicate that these effects result from a dilution effect rather than a true decrease in total capacity; the tissue may thus be said to be in a state of pseudo-atrophy. However, under conditions of very low physiological stimulation of BAT, glucocorticoids may truly inhibit Ucp1 gene expression and consequently lower total UCP1 protein levels, but the metabolic effects of this reduction are probably minor. It is thus unlikely that glucocorticoids affect organismal metabolism and induce the development of obesity through alterations of BAT activity.
Collapse
Affiliation(s)
- Ineke H N Luijten
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
43
|
Fifty shades of brown: The functions, diverse regulation and evolution of brown adipose tissue. Mol Aspects Med 2019; 68:1-5. [PMID: 31325457 DOI: 10.1016/j.mam.2019.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|