1
|
Zhang Y, Ma T, Lu X, Hua H, Wu L, Chen Z. Mechanical mechanics-reclaiming a new battlefield for chronic liver disease. J Adv Res 2025:S2090-1232(25)00346-7. [PMID: 40379238 DOI: 10.1016/j.jare.2025.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/17/2025] [Accepted: 05/12/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND In the 21st century, significant breakthroughs have been made in the research of chronic liver disease. New biochemical markers of pathogenicity and corresponding drugs continue to emerge. However, current treatment strategies remain unsatisfactory due to complex pathological changes in the liver, including vascular dysfunction, myofibroblast-like transition, and local tissue necrosis in liver sinusoids. These challenges have created an urgent need for innovative, synergistic treatments. Mechanical mechanics is a growing field, with increasing evidence suggesting that mechanical signals play a role similar to that of biochemical markers. These signals influence response speed, conduction intensity, and functional diversity in regulating cell activities. AIM OF REVIEW This review summarizes the three main mechanical characteristics involved in the progression of "liver fibrosis-cirrhosis-hepatocellular carcinoma" and provides an in-depth interpretation of several mechanically-related targets. Finally, current and cutting-edge therapeutic strategies are proposed from a cellular perspective. Despite the many challenges that remain, this review is both relevant and significant.
Collapse
Affiliation(s)
- Yiheng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Tianle Ma
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - XingXing Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Haibing Hua
- Department of Gastroenterology, Jiangyin Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Jiangyin 214400, China.
| | - Li Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Zhipeng Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Lecoutre S, Rebière C, Maqdasy S, Lambert M, Dussaud S, Abatan JB, Dugail I, Gautier EL, Clément K, Marcelin G. Enhancing adipose tissue plasticity: progenitor cell roles in metabolic health. Nat Rev Endocrinol 2025; 21:272-288. [PMID: 39757324 DOI: 10.1038/s41574-024-01071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 01/07/2025]
Abstract
Adipose tissue demonstrates considerable plasticity and heterogeneity, enabling metabolic, cellular and structural adaptations to environmental signals. This adaptability is key for maintaining metabolic homeostasis. Impaired adipose tissue plasticity can lead to abnormal adipose tissue responses to metabolic cues, which contributes to the development of cardiometabolic diseases. In chronic obesity, white adipose tissue undergoes pathological remodelling marked by adipocyte hypertrophy, chronic inflammation and fibrosis, which are linked to local and systemic insulin resistance. Research data suggest that the capacity for healthy or unhealthy white adipose tissue remodelling might depend on the intrinsic diversity of adipose progenitor cells (APCs), which sense and respond to metabolic cues. This Review highlights studies on APCs as key determinants of adipose tissue plasticity, discussing differences between subcutaneous and visceral adipose tissue depots during development, growth and obesity. Modulating APC functions could improve strategies for treating adipose tissue dysfunction and metabolic diseases in obesity.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| | - Clémentine Rebière
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Salwan Maqdasy
- Department of Medicine, Karolinska Institutet Hospital, Stockholm, Sweden
| | - Mélanie Lambert
- Institut National de la Santé et de la Recherche Médicale, Bobigny, France
- Labex Inflamex, Université Sorbonne Paris Nord, Alliance Sorbonne Paris Cité, Bobigny, France
| | - Sébastien Dussaud
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Jimon Boniface Abatan
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Emmanuel L Gautier
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
- Department of Nutrition, Pitie-Salpêtriere Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Geneviève Marcelin
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| |
Collapse
|
3
|
Neira G, Hernández-Pardos AW, Becerril S, Ramírez B, Valentí V, Moncada R, Catalán V, Gómez-Ambrosi J, Burrell MA, Silva C, Escalada J, Frühbeck G, Rodríguez A. Differential mitochondrial adaptation and FNDC5 production in brown and white adipose tissue in response to cold and obesity. Obesity (Silver Spring) 2024; 32:2120-2134. [PMID: 39327772 DOI: 10.1002/oby.24132] [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: 04/24/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 09/28/2024]
Abstract
OBJECTIVE Fibronectin type III domain-containing protein 5 (FNDC5) modulates adipocyte metabolism by increasing white and brown adipose tissue (WAT and BAT) browning and activity, respectively. We investigated whether FNDC5 can regulate visceral WAT and BAT adaptive thermogenesis by improving mitochondrial homeostasis in response to cold and obesity. METHODS Adipose tissue expression of FNDC5 and factors involved in mitochondrial homeostasis were determined in patients with normal weight and obesity (n = 159) and in rats with diet-induced obesity after 1 week of cold exposure (n = 61). The effect of different FNDC5 concentrations on mitochondrial biogenesis, dynamics, and mitophagy was evaluated in vitro in human adipocytes. RESULTS In human visceral adipocytes, FNDC5/irisin triggered mitochondrial biogenesis (TFAM) and fusion (MFN1, MFN2, and OPA1) while inhibiting peripheral fission (DNM1L and FIS1) and mitophagy (PINK1 and PRKN). Circulating and visceral WAT expression of FNDC5 was decreased in patients and experimental animals with obesity, whereas its receptor, integrin αV, was upregulated. Obesity increased mitochondrial fusion while decreasing mitophagy in visceral WAT from patients and rats. By contrast, in rat BAT, an upregulation of Fndc5 and genes involved in mitochondrial biogenesis and fission was observed. Cold exposure promoted mitochondrial biogenesis and healthy peripheral fission while repressing Fndc5 expression and mitophagy in BAT from rats. CONCLUSIONS Depot differences in FNDC5 production and mitochondrial adaptations in response to obesity and cold might indicate a self-regulatory mechanism to control thermogenesis in response to energy needs.
Collapse
Affiliation(s)
- Gabriela Neira
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Víctor Valentí
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - María A Burrell
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Camilo Silva
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Escalada
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
4
|
Bahn YJ, Wang Y, Dagur P, Scott N, Cero C, Long KT, Nguyen N, Cypess AM, Rane SG. TGF-β antagonism synergizes with PPARγ agonism to reduce fibrosis and enhance beige adipogenesis. Mol Metab 2024; 90:102054. [PMID: 39461664 PMCID: PMC11570741 DOI: 10.1016/j.molmet.2024.102054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
OBJECTIVES Adipose tissue depots vary markedly in their ability to store and metabolize triglycerides, undergo beige adipogenesis and susceptibility to metabolic disease. The molecular mechanisms that underlie such heterogeneity are not entirely clear. Previously, we showed that TGF-β signaling suppresses beige adipogenesis via repressing the recruitment of dedicated beige progenitors. Here, we find that TGF-β signals dynamically regulate the balance between adipose tissue fibrosis and beige adipogenesis. METHODS We investigated adipose tissue depot-specific differences in activation of TGF-β signaling in response to dietary challenge. RNA-seq and fluorescence activated cell sorting was performed to identify and characterize cells responding to changes in TGF-β signaling status. Mouse models, pharmacological strategies and human adipose tissue analyses were performed to further define the influence of TGF-β signaling on fibrosis and functional beige adipogenesis. RESULTS Elevated basal and high-fat diet inducible activation of TGF-β/Smad3 signaling was observed in the visceral adipose tissue depot. Activation of TGF-β/Smad3 signaling was associated with increased adipose tissue fibrosis. RNA-seq combined with fluorescence-activated cell sorting of stromal vascular fraction of epididymal white adipose tissue depot resulted in identification of TGF-β/Smad3 regulated ITGA5+ fibrogenic progenitors. TGF-β/Smad3 signal inhibition, genetically or pharmacologically, reduced fibrosis and increased functional beige adipogenesis. TGF-β/Smad3 antagonized the beneficial effects of PPARγ whereas TGF-β receptor 1 inhibition synergized with actions of rosiglitazone, a PPARγ agonist, to dampen fibrosis and promote beige adipogenesis. Positive correlation between TGF-β activation and ITGA5 was observed in human adipose tissue, with visceral adipose tissue depots exhibiting higher fibrosis potential than subcutaneous or brown adipose tissue depots. CONCLUSIONS Basal and high-fat diet inducible activation of TGF-β underlies the heterogeneity of adipose tissue depots. TGF-β/Smad3 activation promotes adipose tissue fibrosis and suppresses beige progenitors. Together, these dual mechanisms preclude functional beige adipogenesis. Controlled inhibition of TβRI signaling and concomitant PPARγ stimulation can suppress adipose tissue fibrosis and promote beige adipogenesis to improve metabolism.
Collapse
Affiliation(s)
- Young Jae Bahn
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Yanling Wang
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Pradeep Dagur
- Flow Cytometry Core, National Heart, Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Nicholas Scott
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Cheryl Cero
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Kelly T Long
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Nhuquynh Nguyen
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Sushil G Rane
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.
| |
Collapse
|
5
|
Yan XY, Luo YY, Chen HJ, Hu XQ, Zheng P, Fang HT, Ding F, Zhang L, Li Z, Yan YE. IRX3 promotes adipose tissue browning and inhibits fibrosis in obesity-resistant mice. Int J Biochem Cell Biol 2024; 175:106638. [PMID: 39173825 DOI: 10.1016/j.biocel.2024.106638] [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: 01/11/2024] [Revised: 08/05/2024] [Accepted: 08/17/2024] [Indexed: 08/24/2024]
Abstract
Obesity is one of the threats to human health and survival. High fat diet (HFD)-induced obesity leads to adipose tissue fibrosis and a series of metabolic diseases. There are some people still thin under HFD, a phenomenon known as the "obesity resistance (OR) phenotype". It was found that Iroquois homeobox 3 (IRX3) is considered as a regulator in obesity, but the regulatory mechanism between OR and IRX3 is still unclear. In this study, we investigated OR on a HFD and the role of the IRX3 gene. Using mice, we observed that OR mice had lower body weights, reduced liver lipid synthesis, and increased white adipose tissue (WAT) lipolysis compared to obesity-prone (OP) mice. Additionally, OR mice exhibited spontaneous WAT browning and less fibrosis, correlating with higher Irx3 expression. Utilizing 3T3-L1 differentiated adipocytes, our study demonstrated that overexpression of Irx3 promoted thermogenesis-related gene expression and reduced adipocyte fibrosis. Therefore, Irx3 promotes WAT browning and inhibits fibrosis in OR mice. These results provide insight into the differences between obesity and OR, new perspectives on obesity treatment, and guidance for lessening adipose tissue fibrosis.
Collapse
Affiliation(s)
- Xi-Yue Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Yuan-Yuan Luo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hui-Jian Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Xiao-Qin Hu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Peng Zheng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hong-Ting Fang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Fei Ding
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Li Zhang
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Zhen Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China.
| | - You-E Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
| |
Collapse
|
6
|
Zhang Q, Lu C, Lu F, Liao Y, Cai J, Gao J. Challenges and opportunities in obesity: the role of adipocytes during tissue fibrosis. Front Endocrinol (Lausanne) 2024; 15:1365156. [PMID: 38686209 PMCID: PMC11056552 DOI: 10.3389/fendo.2024.1365156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Obesity is a chronic disease that affects the energy balance of the whole body. In addition to increasing fat mass, tissue fibrosis occurred in white adipose tissue in obese condition. Fibrosis is the over-activation of fibroblasts leading to excessive accumulation of extracellular matrix, which could be caused by various factors, including the status of adipocytes. The morphology of adipocytes responds rapidly and dynamically to nutrient fluctuations. Adaptive hypertrophy of normal adipocytes protects peripheral organs from damage from lipotoxicity. However, the biological behavior of hypertrophic adipocytes in chronic obesity is abnormally altered. Adipocytes lead to fibrotic remodeling of the extracellular matrix by inducing unresolved chronic inflammation, persistent hypoxia, and increasing myofibroblast numbers. Moreover, adipocyte-induced fibrosis not only restricts the flexible expansion and contraction of adipose tissue but also initiates the development of various diseases through cellular autonomic and paracrine effects. Regarding anti-fibrotic therapy, dysregulated intracellular signaling and epigenetic changes represent potential candidate targets. Thus, modulation of adipocytes may provide potential therapeutic avenues for reversing pathological fibrosis in adipose tissue and achieving the anti-obesity purpose.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chongxuan Lu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junrong Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Zhao JY, Zhou LJ, Ma KL, Hao R, Li M. MHO or MUO? White adipose tissue remodeling. Obes Rev 2024; 25:e13691. [PMID: 38186200 DOI: 10.1111/obr.13691] [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: 05/05/2023] [Revised: 11/14/2023] [Accepted: 11/19/2023] [Indexed: 01/09/2024]
Abstract
In this review, we delve into the intricate relationship between white adipose tissue (WAT) remodeling and metabolic aspects in obesity, with a specific focus on individuals with metabolically healthy obesity (MHO) and metabolically unhealthy obesity (MUO). WAT is a highly heterogeneous, plastic, and dynamically secreting endocrine and immune organ. WAT remodeling plays a crucial role in metabolic health, involving expansion mode, microenvironment, phenotype, and distribution. In individuals with MHO, WAT remodeling is beneficial, reducing ectopic fat deposition and insulin resistance (IR) through mechanisms like increased adipocyte hyperplasia, anti-inflammatory microenvironment, appropriate extracellular matrix (ECM) remodeling, appropriate vascularization, enhanced WAT browning, and subcutaneous adipose tissue (SWAT) deposition. Conversely, for those with MUO, WAT remodeling leads to ectopic fat deposition and IR, causing metabolic dysregulation. This process involves adipocyte hypertrophy, disrupted vascularization, heightened pro-inflammatory microenvironment, enhanced brown adipose tissue (BAT) whitening, and accumulation of visceral adipose tissue (VWAT) deposition. The review underscores the pivotal importance of intervening in WAT remodeling to hinder the transition from MHO to MUO. This insight is valuable for tailoring personalized and effective management strategies for patients with obesity in clinical practice.
Collapse
Affiliation(s)
- Jing Yi Zhao
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Juan Zhou
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kai Le Ma
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Hao
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Li
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Eisinger K, Girke P, Buechler C, Krautbauer S. Adipose tissue depot specific expression and regulation of fibrosis-related genes and proteins in experimental obesity. Mamm Genome 2024; 35:13-30. [PMID: 37884762 PMCID: PMC10884164 DOI: 10.1007/s00335-023-10022-3] [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: 10/14/2022] [Accepted: 10/02/2023] [Indexed: 10/28/2023]
Abstract
Transforming growth factor beta (Tgfb) is a well-studied pro-fibrotic cytokine, which upregulates cellular communication network factor 2 (Ccn2), collagen, and actin alpha 2, smooth muscle (Acta2) expression. Obesity induces adipose tissue fibrosis, which contributes to metabolic diseases. This work aimed to analyze the expression of Tgfb, Ccn2, collagen1a1 (Col1a1), Acta2 and BMP and activin membrane-bound inhibitor (Bambi), which is a negative regulator of Tgfb signaling, in different adipose tissue depots of mice fed a standard chow, mice fed a high fat diet (HFD) and ob/ob mice. Principally, these genes were low expressed in brown adipose tissues and this difference was less evident for the ob/ob mice. Ccn2 and Bambi protein as well as mRNA expression, and collagen1a1 mRNA were not induced in the adipose tissues upon HFD feeding whereas Tgfb and Acta2 mRNA increased in the white fat depots. Immunoblot analysis showed that Acta2 protein was higher in subcutaneous and perirenal fat of these mice. In the ob/ob mice, Ccn2 mRNA and Ccn2 protein were upregulated in the fat depots. Here, Tgfb, Acta2 and Col1a1 mRNA levels and serum Tgfb protein were increased. Acta2 protein was, however, not higher in subcutaneous and perirenal fat of these mice. Col6a1 mRNA was shown before to be higher in obese fat tissues. Current analysis proved the Col6a1 protein was induced in subcutaneous fat of HFD fed mice. Notably, Col6a1 was reduced in perirenal fat of ob/ob mice in comparison to the respective controls. 3T3-L1 cells express Ccn2 and Bambi protein, whose levels were not changed by fatty acids, leptin, lipopolysaccharide, tumor necrosis factor and interleukin-6. All of these factors led to higher Tgfb in 3T3-L1 adipocyte media but did not increase its mRNA levels. Free fatty acids induced necrosis whereas apoptosis did not occur in any of the in vitro incubations excluding cell death as a main reason for higher Tgfb in cell media. In summary, Tgfb mRNA is consistently induced in white fat tissues in obesity but this is not paralleled by a clear increase of its target genes. Moreover, discrepancies between mRNA and protein expression of Acta2 were observed. Adipocytes seemingly do not contribute to higher Tgfb mRNA levels in obesity. These cells release more Tgfb protein when challenged with obesity-related metabolites connecting metabolic dysfunction and fibrosis.
Collapse
Affiliation(s)
- Kristina Eisinger
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Philipp Girke
- Department of Genetics, University of Regensburg, 93040, Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany.
| | - Sabrina Krautbauer
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| |
Collapse
|
9
|
Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose Tissue Dysfunction Determines Lipotoxicity and Triggers the Metabolic Syndrome: Current Challenges and Clinical Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:231-272. [PMID: 39287854 DOI: 10.1007/978-3-031-63657-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis, and it is constituted of three different types of adipocytes: white, beige and brown, which are integrated with vascular, immune, neural, and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concerted action of the three types of adipocytes/tissues ensures an optimal metabolic status. However, when one or several of these adipose depots become dysfunctional because of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations close a vicious cycle that negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and ensuring its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity are complementary strategies that counteract obesity and its associated lipotoxic metabolic effects. However, the development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter, we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition, and expandability capacity potential as well as molecular and metabolic characteristic signatures in both physiological and pathophysiological conditions. Current antilipotoxic strategies for future clinical application are also discussed in this chapter.
Collapse
Affiliation(s)
- Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| | - Vanessa Pellegrinelli
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| |
Collapse
|
10
|
Abstract
Adipose tissue exhibits a remarkable capacity to expand, contract, and remodel in response to changes in physiological and environmental conditions. Here, we describe recent advances in our understanding of how functionally distinct tissue-resident mesenchymal stromal cell subpopulations orchestrate several aspects of physiological and pathophysiological adipose tissue remodeling, with a particular focus on the adaptations that occur in response to changes in energy surplus and environmental temperature. The study of adipose tissue remodeling provides a vehicle to understand the functional diversity of stromal cells and offers a lens through which several generalizable aspects of tissue reorganization can be readily observed.
Collapse
Affiliation(s)
- Jessica Cannavino
- Department of Medicine, Division of Endocrinology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina 27701, USA
| | - Rana K Gupta
- Department of Medicine, Division of Endocrinology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina 27701, USA
| |
Collapse
|
11
|
Jääskeläinen I, Petäistö T, Mirzarazi Dahagi E, Mahmoodi M, Pihlajaniemi T, Kaartinen MT, Heljasvaara R. Collagens Regulating Adipose Tissue Formation and Functions. Biomedicines 2023; 11:biomedicines11051412. [PMID: 37239083 DOI: 10.3390/biomedicines11051412] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
The globally increasing prevalence of obesity is associated with the development of metabolic diseases such as type 2 diabetes, dyslipidemia, and fatty liver. Excess adipose tissue (AT) often leads to its malfunction and to a systemic metabolic dysfunction because, in addition to storing lipids, AT is an active endocrine system. Adipocytes are embedded in a unique extracellular matrix (ECM), which provides structural support to the cells as well as participating in the regulation of their functions, such as proliferation and differentiation. Adipocytes have a thin pericellular layer of a specialized ECM, referred to as the basement membrane (BM), which is an important functional unit that lies between cells and tissue stroma. Collagens form a major group of proteins in the ECM, and some of them, especially the BM-associated collagens, support AT functions and participate in the regulation of adipocyte differentiation. In pathological conditions such as obesity, AT often proceeds to fibrosis, characterized by the accumulation of large collagen bundles, which disturbs the natural functions of the AT. In this review, we summarize the current knowledge on the vertebrate collagens that are important for AT development and function and include basic information on some other important ECM components, principally fibronectin, of the AT. We also briefly discuss the function of AT collagens in certain metabolic diseases in which they have been shown to play central roles.
Collapse
Affiliation(s)
- Iida Jääskeläinen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Tiina Petäistö
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Elahe Mirzarazi Dahagi
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Mahdokht Mahmoodi
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Taina Pihlajaniemi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Mari T Kaartinen
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Ritva Heljasvaara
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| |
Collapse
|
12
|
Xiong X, Li W, Liu R, Saha P, Yechoor V, Ma K. Circadian clock control of MRTF/SRF pathway suppresses beige adipocyte thermogenic recruitment. J Mol Cell Biol 2023; 14:mjac079. [PMID: 36581314 PMCID: PMC10174720 DOI: 10.1093/jmcb/mjac079] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/21/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
The morphological transformation of adipogenic progenitors into mature adipocytes requires dissolution of actin cytoskeleton with loss of myocardin-related transcription factor (MRTF)/serum response factor (SRF) activity. Circadian clock confers temporal control in adipogenic differentiation, while the actin cytoskeleton-MRTF/SRF signaling transduces extracellular physical niche cues. Here, we define a novel circadian transcriptional control involved in actin cytoskeleton-MRTF/SRF signaling cascade that modulates beige fat thermogenic function. Key components of actin dynamic-MRTF/SRF pathway display circadian regulation in beige fat depot. The core clock regulator, brain and muscle arnt-like 1 (Bmal1), exerts direct transcriptional control of genes within the actin dynamic-MRTF/SRF cascade that impacts actin cytoskeleton organization and SRF activity. Employing beige fat-selective gene-targeting models together with pharmacological rescues, we further demonstrate that Bmal1 inhibits beige adipogenesis and thermogenic capacity in vivo via the MRTF/SRF pathway. Selective ablation of Bmal1 induces beigeing with improved glucose homeostasis, whereas its targeted overexpression attenuates thermogenic induction resulting in obesity. Collectively, our findings identify the clock-MRTF/SRF regulatory axis as an inhibitory mechanism of beige fat thermogenic recruitment with significant contribution to systemic metabolic homeostasis.
Collapse
Affiliation(s)
- Xuekai Xiong
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Weini Li
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Ruya Liu
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Pradip Saha
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Vijay Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ke Ma
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
13
|
Cai Z, He B. Adipose tissue aging: An update on mechanisms and therapeutic strategies. Metabolism 2023; 138:155328. [PMID: 36202221 DOI: 10.1016/j.metabol.2022.155328] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/20/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
Aging is a complex biological process characterized by a progressive loss of physiological integrity and increased vulnerability to age-related diseases. Adipose tissue plays central roles in the maintenance of whole-body metabolism homeostasis and has recently attracted significant attention as a biological driver of aging and age-related diseases. Here, we review the most recent advances in our understanding of the molecular and cellular mechanisms underlying age-related decline in adipose tissue function. In particular, we focus on the complex inter-relationship between metabolism, immune, and sympathetic nervous system within adipose tissue during aging. Moreover, we discuss the rejuvenation strategies to delay aging and extend lifespan, including senescent cell ablation (senolytics), dietary intervention, physical exercise, and heterochronic parabiosis. Understanding the pathological mechanisms that underlie adipose tissue aging will be critical for the development of new intervention strategies to slow or reverse aging and age-related diseases.
Collapse
Affiliation(s)
- Zhaohua Cai
- Heart Center, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Ben He
- Heart Center, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China.
| |
Collapse
|
14
|
Hu B, Shen X, Qin W, Zhang L, Zou T, Dong Q, Qin LX. A Prognostic Nomogram for Hepatocellular Carcinoma Based on Wound Healing and Immune Checkpoint Genes. J Clin Transl Hepatol 2022; 10:891-900. [PMID: 36304515 PMCID: PMC9547254 DOI: 10.14218/jcth.2021.00296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/18/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND AIMS Wound healing and tumor progression share some common biological features; however, how variations in wound healing patterns affect hepatocellular carcinoma (HCC) prognosis remains unclear. METHODS We analyzed the wound healing patterns of 594 HCC samples from The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC) and correlated them with immune infiltration and the expression levels of immune checkpoint genes. A risk score, which we named the "heal.immune" score, was established via stepwise Cox estimation. We constructed a nomogram based on age, sex, TNM stage, and heal.immune score and explored its predictive value for HCC prognosis. Seventy-four clinical patients were enrolled in this study, and all were from Huashan Hospital of Fudan University between 2015 and 2017 to serve as an independent validation group. RESULTS We identified two distinct wound healing patterns in HCC. The biological processes of healing cluster 1 (C1) are related to metabolism, while those of healing cluster 2 (C2) are related to the inflammatory response and immune cell accumulation. A total of 565 wound healing-related genes (based on Gene Ontology) and 25 immune checkpoint genes were considered. By analyzing differentially expressed genes and implementing a stepwise Cox estimation analysis, six genes with p values less than 0.02 in a multivariate Cox estimation were chosen as the "heal.immune" gene set (FCER1G, PLAT, ITGA5, CCNB1, CD86 and CD40). The "heal.immune" gene set, as an OS risk factor, was further validated in Fudan cohort. We constructed a nomogram to predict the 1-, 3- and 5-year overall survival (OS) in the TCGA cohort. The area under curve vales of the receiver characteristic operator curves were 0.82, 0.76 and 0.73 in the training group and 0.84, 0.76 and 0.72 in the test group. CONCLUSIONS We established a prognostic nomogram based on the heal.immune gene signature, which includes six wound healing- and immunity-related genes. This nomogram accurately predicts the OS of HCC patients.
Collapse
Affiliation(s)
| | | | | | - Lan Zhang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tiantian Zou
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qiongzhu Dong
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
15
|
The Hippo pathway links adipocyte plasticity to adipose tissue fibrosis. Nat Commun 2022; 13:6030. [PMID: 36229481 PMCID: PMC9562301 DOI: 10.1038/s41467-022-33800-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2022] Open
Abstract
Fibrosis disrupts adipose tissue (AT) homeostasis and exacerbates metabolic dysfunction upon chronic caloric excess. The molecular mechanisms linking adipocyte plasticity to AT fibrosis are largely unknown. Here we show that the Hippo pathway is coupled with TGFβ signaling to orchestrate a cellular and/or functional shift of adipocytes from energy storage to extracellular matrix (ECM) remodeling in AT fibrosis. We found that Lats1/2-knockout adipocytes could dedifferentiate into DPP4+ progenitor cells and convert to DPP4− myofibroblasts upon TGFβ stimulation. On the other hand, Hippo pathway inhibition during obesity impaired adipocyte identity while promoted ECM remodeling activity of adipocytes. Macrophages recruited by CCL2 produced TGFβ to accelerate AT fibrosis. YAP and TAZ, the Hippo downstream effectors, enhanced SMAD2 stability to promote fibrotic responses. Importantly, inhibition of YAP/TAZ activity in obese mice markedly relieved AT fibrosis and improved metabolic homeostasis. Together, our findings identify the Hippo pathway as a molecular switch in the initiation and development of AT fibrosis, implying it as a therapeutic target. Adipose tissue fibrosis is connected to obesity-related metabolic dysfunction. Qiu and colleagues discover that the Hippo pathway acts as a molecular switch in the initiation and development of adipose tissue fibrosis upon TGFβ stimulation.
Collapse
|
16
|
Xi Y, LaCanna R, Ma HY, N'Diaye EN, Gierke S, Caplazi P, Sagolla M, Huang Z, Lucio L, Arlantico A, Jeet S, Brightbill H, Emson C, Wong A, Morshead KB, DePianto DJ, Roose-Girma M, Yu C, Tam L, Jia G, Ramalingam TR, Marsters S, Ashkenazi A, Kim SH, Kelly R, Wu S, Wolters PJ, Feldstein AE, Vander Heiden JA, Ding N. A WISP1 antibody inhibits MRTF signaling to prevent the progression of established liver fibrosis. Cell Metab 2022; 34:1377-1393.e8. [PMID: 35987202 DOI: 10.1016/j.cmet.2022.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 06/06/2022] [Accepted: 07/19/2022] [Indexed: 01/18/2023]
Abstract
Fibrosis is the major risk factor associated with morbidity and mortality in patients with non-alcoholic steatohepatitis (NASH)-driven chronic liver disease. Although numerous efforts have been made to identify the mediators of the initiation of liver fibrosis, the molecular underpinnings of fibrosis progression remain poorly understood, and therapies to arrest liver fibrosis progression are elusive. Here, we identify a pathway involving WNT1-inducible signaling pathway protein 1 (WISP1) and myocardin-related transcription factor (MRTF) as a central mechanism driving liver fibrosis progression through the integrin-dependent transcriptional reprogramming of myofibroblast cytoskeleton and motility. In mice, WISP1 deficiency protects against fibrosis progression, but not fibrosis onset. Moreover, the therapeutic administration of a novel antibody blocking WISP1 halted the progression of existing liver fibrosis in NASH models. These findings implicate the WISP1-MRTF axis as a crucial determinant of liver fibrosis progression and support targeting this pathway by antibody-based therapy for the treatment of NASH fibrosis.
Collapse
Affiliation(s)
- Ying Xi
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Ryan LaCanna
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Hsiao-Yen Ma
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Elsa-Noah N'Diaye
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Sarah Gierke
- Department of Pathology, Genentech, South San Francisco, CA, USA
| | - Patrick Caplazi
- Department of Pathology, Genentech, South San Francisco, CA, USA
| | - Meredith Sagolla
- Department of Pathology, Genentech, South San Francisco, CA, USA
| | - Zhiyu Huang
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Laura Lucio
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Alexander Arlantico
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Surinder Jeet
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Hans Brightbill
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Claire Emson
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Aaron Wong
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Katrina B Morshead
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Daryle J DePianto
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Merone Roose-Girma
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Charles Yu
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Lucinda Tam
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Guiquan Jia
- Department of Biomarker Discovery, Genentech, South San Francisco, CA, USA
| | | | - Scot Marsters
- Department of Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Avi Ashkenazi
- Department of Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Si Hyun Kim
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Ryan Kelly
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Shuang Wu
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Paul J Wolters
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ariel E Feldstein
- Department of Pediatrics, University of California, San Diego, San Diego, CA, USA
| | | | - Ning Ding
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
17
|
Xiong S, Tan J, Wang Y, He J, Hu F, Wu X, Liu Z, Lin S, Li X, Chen Z, Mao R. Fibrosis in fat: From other diseases to Crohn’s disease. Front Immunol 2022; 13:935275. [PMID: 36091035 PMCID: PMC9453038 DOI: 10.3389/fimmu.2022.935275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
Creeping fat is a specific feature of Crohn’s disease (CD) and is characterized by mesenteric fat wrapping around the intestine. It highly correlates with intestinal transmural inflammation, muscular hypertrophy, fibrosis, and stricture formation. However, the pathogenesis of creeping fat remains unclear. Molecular crosstalk exists between mesenteric fat and the intestine. Indeed, creeping fat contains different types of cells, including adipocytes and immune cells. These cell types can produce various cytokines, fatty acids, and growth factors, which affect the mesenteric fat function and modulate intestinal inflammation and immunity. Moreover, adipocyte progenitors can produce extracellular matrix to adapt to fat expansion. Previous studies have shown that fat fibrosis is an important feature of adipose tissue malfunction and exists in other diseases, including metabolic disorders, cancer, atrial fibrillation, and osteoarthritis. Furthermore, histological sections of CD showed fibrosis in the creeping fat. However, the role of fibrosis in the mesenteric fat of CD is not well understood. In this review, we summarized the possible mechanisms of fat fibrosis and its impact on other diseases. More specifically, we illustrated the role of various cells (adipocyte progenitors, macrophages, mast cells, and group 1 innate lymphoid cells) and molecules (including hypoxia-inducible factor 1-alpha, transforming growth factor-beta, platelet-derived growth factor, and peroxisome proliferator-activated receptor-gamma) in the pathogenesis of fat fibrosis in other diseases to understand the role of creeping fat fibrosis in CD pathogenesis. Future research will provide key information to decipher the role of fat fibrosis in creeping fat formation and intestinal damage, thereby helping us identify novel targets for the diagnosis and treatment of CD.
Collapse
Affiliation(s)
- Shanshan Xiong
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jinyu Tan
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jinshen He
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fan Hu
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaomin Wu
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zishan Liu
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sinan Lin
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuehua Li
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhihui Chen
- Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Ren Mao, ; Zhihui Chen,
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Gastroenterology, Huidong People’s Hospital, Huizhou, China
- *Correspondence: Ren Mao, ; Zhihui Chen,
| |
Collapse
|
18
|
Abstract
Adipose tissue is a complex heterogeneous tissue composed of adipocytes along with several non-adipocyte populations, including blood, stromal, endothelial, and progenitor cells, as well as extracellular matrix (ECM) components. As obesity progresses, the adipose tissue expands dynamically through adipocyte hypertrophy and/or hyperplasia. This expansion requires continuous ECM remodeling to properly accommodate the size increase as well as functional changes. Upon reaching a hypertrophic threshold beyond the adipocyte buffering capacity, excess ECM components are deposited, causing fibrosis and ultimately resulting in unhealthy metabolic maladaptation. These complex ECM remodeling processes in adipose tissues are regulated by the local environment, several key mediators, and genetic factors that are closely linked to insulin sensitivity. It is crucial to understand how adipocytes interact with nonadipocyte populations and various mediators (i.e., immune cells, ECM components, and adipokines) during these processes. This mini-review provides an overview of the latest research into the biology of obesity-induced adipose tissue fibrosis and its related clinical manifestations, providing insight for further studies aimed at controlling metabolic syndrome and its comorbidities.
Collapse
Affiliation(s)
- Yutaka Hasegawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| |
Collapse
|
19
|
Aissa AF, Tryndyak VP, de Conti A, Rita Thomazela Machado A, Tuttis K, da Silva Machado C, Hernandes LC, Wellington da Silva Santos P, Mara Serpeloni J, P Pogribny I, Maria Greggi Antunes L. Epigenetic changes induced in mice liver by methionine-supplemented and methionine-deficient diets. Food Chem Toxicol 2022; 163:112938. [PMID: 35314295 DOI: 10.1016/j.fct.2022.112938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 02/07/2023]
Abstract
A diet deficient in donors of methyl group, such as methionine, affects DNA methylation and hepatic lipid metabolism. Methionine also affects other epigenetic mechanisms, such as microRNAs. We investigated the effects of methionine-supplemented or methionine-deficient diets on the expression of chromatin-modifying genes, global DNA methylation, the expression and methylation of genes related to lipid metabolism, and the expression of microRNAs in mouse liver. Female Swiss albino mice were fed a control diet (0.3% methionine), a methionine-supplemented diet (2% methionine), and a methionine-deficient diet (0% methionine) for 10 weeks. The genes most affected by the methionine-supplemented diet were associated with histone and DNA methyltransferases activity, while the methionine-deficient diet mostly altered the expression of histone methyltransferases genes. Both diets altered the global DNA methylation and the expression and gene-specific methylation of the lipid metabolism gene Apoa5. Both diets altered the expression of several liver homeostasis-related microRNAs, including miR-190b-5p, miR-130b-3p, miR-376c-3p, miR-411-5p, miR-29c-3p, miR-295-3p, and miR-467d-5p, with the methionine-deficient diet causing a more substantial effect. The effects of improper amounts of methionine in the diet on liver pathologies may involve a cooperative action of chromatin-modifying genes, which results in an aberrant pattern of global and gene-specific methylation, and microRNAs responsible for liver homeostasis.
Collapse
Affiliation(s)
- Alexandre Ferro Aissa
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Volodymyr P Tryndyak
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Aline de Conti
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Ana Rita Thomazela Machado
- Departament of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Katiuska Tuttis
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Carla da Silva Machado
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Lívia Cristina Hernandes
- Departament of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Patrick Wellington da Silva Santos
- Departament of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Juliana Mara Serpeloni
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina, PR, Brazil
| | - Igor P Pogribny
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Lusânia Maria Greggi Antunes
- Departament of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
20
|
Jansen J, Reimer KC, Nagai JS, Varghese FS, Overheul GJ, de Beer M, Roverts R, Daviran D, Fermin LA, Willemsen B, Beukenboom M, Djudjaj S, von Stillfried S, van Eijk LE, Mastik M, Bulthuis M, Dunnen WD, van Goor H, Hillebrands JL, Triana SH, Alexandrov T, Timm MC, van den Berge BT, van den Broek M, Nlandu Q, Heijnert J, Bindels EM, Hoogenboezem RM, Mooren F, Kuppe C, Miesen P, Grünberg K, Ijzermans T, Steenbergen EJ, Czogalla J, Schreuder MF, Sommerdijk N, Akiva A, Boor P, Puelles VG, Floege J, Huber TB, van Rij RP, Costa IG, Schneider RK, Smeets B, Kramann R, Achdout H, Aimon A, Bar-David E, Barr H, Ben-Shmuel A, Bennett J, Boby ML, Borden B, Bowman GR, Brun J, BVNBS S, Calmiano M, Carbery A, Cattermole E, Chernychenko E, Choder JD, Clyde A, Coffland JE, Cohen G, Cole J, Contini A, Cox L, Cvitkovic M, Dias A, Donckers K, Dotson DL, Douangamath A, Duberstein S, Dudgeon T, Dunnett L, Eastman PK, Erez N, Eyermann CJ, Fairhead M, Fate G, Fearon D, Federov O, Ferla M, Fernandes RS, Ferrins L, Foster R, Foster H, Gabizon R, Garcia-Sastre A, Gawriljuk VO, Gehrtz P, Gileadi C, Giroud C, Glass WG, Glen R, Itai glinert, Godoy AS, Gorichko M, et alJansen J, Reimer KC, Nagai JS, Varghese FS, Overheul GJ, de Beer M, Roverts R, Daviran D, Fermin LA, Willemsen B, Beukenboom M, Djudjaj S, von Stillfried S, van Eijk LE, Mastik M, Bulthuis M, Dunnen WD, van Goor H, Hillebrands JL, Triana SH, Alexandrov T, Timm MC, van den Berge BT, van den Broek M, Nlandu Q, Heijnert J, Bindels EM, Hoogenboezem RM, Mooren F, Kuppe C, Miesen P, Grünberg K, Ijzermans T, Steenbergen EJ, Czogalla J, Schreuder MF, Sommerdijk N, Akiva A, Boor P, Puelles VG, Floege J, Huber TB, van Rij RP, Costa IG, Schneider RK, Smeets B, Kramann R, Achdout H, Aimon A, Bar-David E, Barr H, Ben-Shmuel A, Bennett J, Boby ML, Borden B, Bowman GR, Brun J, BVNBS S, Calmiano M, Carbery A, Cattermole E, Chernychenko E, Choder JD, Clyde A, Coffland JE, Cohen G, Cole J, Contini A, Cox L, Cvitkovic M, Dias A, Donckers K, Dotson DL, Douangamath A, Duberstein S, Dudgeon T, Dunnett L, Eastman PK, Erez N, Eyermann CJ, Fairhead M, Fate G, Fearon D, Federov O, Ferla M, Fernandes RS, Ferrins L, Foster R, Foster H, Gabizon R, Garcia-Sastre A, Gawriljuk VO, Gehrtz P, Gileadi C, Giroud C, Glass WG, Glen R, Itai glinert, Godoy AS, Gorichko M, Gorrie-Stone T, Griffen EJ, Hart SH, Heer J, Henry M, Hill M, Horrell S, Hurley MF, Israely T, Jajack A, Jnoff E, Jochmans D, John T, De Jonghe S, Kantsadi AL, Kenny PW, Kiappes J, Koekemoer L, Kovar B, Krojer T, Lee AA, Lefker BA, Levy H, London N, Lukacik P, Macdonald HB, Maclean B, Malla TR, Matviiuk T, McCorkindale W, McGovern BL, Melamed S, Michurin O, Mikolajek H, Milne BF, Morris A, Morris GM, Morwitzer MJ, Moustakas D, Nakamura AM, Neto JB, Neyts J, Nguyen L, Noske GD, Oleinikovas V, Oliva G, Overheul GJ, Owen D, Psenak V, Pai R, Pan J, Paran N, Perry B, Pingle M, Pinjari J, Politi B, Powell A, Puni R, Rangel VL, Reddi RN, Reid SP, Resnick E, Ripka EG, Robinson MC, Robinson RP, Rodriguez-Guerra J, Rosales R, Rufa D, Schofield C, Shafeev M, Shaikh A, Shi J, Shurrush K, Sing S, Sittner A, Skyner R, Smalley A, Smilova MD, Solmesky LJ, Spencer J, Strain-Damarell C, Swamy V, Tamir H, Tennant R, Thompson W, Thompson A, Thompson W, Tomasia S, Tumber A, Vakonakis I, van Rij RP, van Geel L, Varghese FS, Vaschetto M, Vitner EB, Voelz V, Volkamer A, von Delft F, von Delft A, Walsh M, Ward W, Weatherall C, Weiss S, White KM, Wild CF, Wittmann M, Wright N, Yahalom-Ronen Y, Zaidmann D, Zidane H, Zitzmann N. SARS-CoV-2 infects the human kidney and drives fibrosis in kidney organoids. Cell Stem Cell 2022; 29:217-231.e8. [PMID: 35032430 PMCID: PMC8709832 DOI: 10.1016/j.stem.2021.12.010] [Show More Authors] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 09/03/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
Abstract
Kidney failure is frequently observed during and after COVID-19, but it remains elusive whether this is a direct effect of the virus. Here, we report that SARS-CoV-2 directly infects kidney cells and is associated with increased tubule-interstitial kidney fibrosis in patient autopsy samples. To study direct effects of the virus on the kidney independent of systemic effects of COVID-19, we infected human-induced pluripotent stem-cell-derived kidney organoids with SARS-CoV-2. Single-cell RNA sequencing indicated injury and dedifferentiation of infected cells with activation of profibrotic signaling pathways. Importantly, SARS-CoV-2 infection also led to increased collagen 1 protein expression in organoids. A SARS-CoV-2 protease inhibitor was able to ameliorate the infection of kidney cells by SARS-CoV-2. Our results suggest that SARS-CoV-2 can directly infect kidney cells and induce cell injury with subsequent fibrosis. These data could explain both acute kidney injury in COVID-19 patients and the development of chronic kidney disease in long COVID.
Collapse
|
21
|
Wei L, Shi J. Insight Into Rho Kinase Isoforms in Obesity and Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:886534. [PMID: 35769086 PMCID: PMC9234286 DOI: 10.3389/fendo.2022.886534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity and associated complications increasingly jeopardize global health and contribute to the rapidly rising prevalence of type 2 diabetes mellitus and obesity-related diseases. Developing novel methods for the prevention and treatment of excess body adipose tissue expansion can make a significant contribution to public health. Rho kinase is a Rho-associated coiled-coil-containing protein kinase (Rho kinase or ROCK). The ROCK family including ROCK1 and ROCK2 has recently emerged as a potential therapeutic target for the treatment of metabolic disorders. Up-regulated ROCK activity has been involved in the pathogenesis of all aspects of metabolic syndrome including obesity, insulin resistance, dyslipidemia and hypertension. The RhoA/ROCK-mediated actin cytoskeleton dynamics have been implicated in both white and beige adipogenesis. Studies using ROCK pan-inhibitors in animal models of obesity, diabetes, and associated complications have demonstrated beneficial outcomes. Studies via genetically modified animal models further established isoform-specific roles of ROCK in the pathogenesis of metabolic disorders including obesity. However, most reported studies have been focused on ROCK1 activity during the past decade. Due to the progress in developing ROCK2-selective inhibitors in recent years, a growing body of evidence indicates more attention should be devoted towards understanding ROCK2 isoform function in metabolism. Hence, studying individual ROCK isoforms to reveal their specific roles and principal mechanisms in white and beige adipogenesis, insulin sensitivity, energy balancing regulation, and obesity development will facilitate significant breakthroughs for systemic treatment with isoform-selective inhibitors. In this review, we give an overview of ROCK functions in the pathogenesis of obesity and insulin resistance with a particular focus on the current understanding of ROCK isoform signaling in white and beige adipogenesis, obesity and thermogenesis in adipose tissue and other major metabolic organs involved in energy homeostasis regulation.
Collapse
Affiliation(s)
- Lei Wei
- *Correspondence: Lei Wei, ; Jianjian Shi,
| | | |
Collapse
|
22
|
Marcelin G, Clément K. The multifaceted progenitor fates in healthy or unhealthy adipose tissue during obesity. Rev Endocr Metab Disord 2021; 22:1111-1119. [PMID: 34105090 DOI: 10.1007/s11154-021-09662-0] [Citation(s) in RCA: 7] [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] [Accepted: 06/01/2021] [Indexed: 10/21/2022]
Abstract
While obesity is defined as an excessive fat accumulation conferring a risk to metabolic health, increased adipose mass by itself does not fully explain obesity's propensity to promote metabolic alterations. Adipose tissue regulates multiple processes critical for energy homeostasis and its dysfunction favors the development and perpetuation of metabolic diseases. Obesity drives inflammatory leucocyte infiltration in adipose tissue and fibrotic transformation of the fat depots. Both features associate with metabolic alterations such as impaired glucose control and resistance to fat mass loss. In this context, adipose progenitors, an heterogenous resident population of mesenchymal stromal cells, display functions important to shape healthy or unhealthy adipose tissue expansion. We, here, outline the current understanding of adipose progenitor biology in the context of obesity-induced adipose tissue remodeling.
Collapse
Affiliation(s)
- Geneviève Marcelin
- Nutrition and Obesities : Systemic Approaches (NutriOmics, UMRS U1269), Sorbonne Universités, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities : Systemic Approaches (NutriOmics, UMRS U1269), Sorbonne Universités, INSERM, Paris, France.
- Nutrition Department, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, CRNH Ile de France, 75013, Paris, France.
| |
Collapse
|
23
|
Hypoxia induces stress fiber formation in adipocytes in the early stage of obesity. Sci Rep 2021; 11:21473. [PMID: 34728615 PMCID: PMC8563745 DOI: 10.1038/s41598-021-00335-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023] Open
Abstract
In obese adipose tissue (AT), hypertrophic expansion of adipocytes is not matched by new vessel formation, leading to AT hypoxia. As a result, hypoxia inducible factor-1⍺ (HIF-1⍺) accumulates in adipocytes inducing a transcriptional program that upregulates profibrotic genes and biosynthetic enzymes such as lysyl oxidase (LOX) synthesis. This excess synthesis and crosslinking of extracellular matrix (ECM) components cause AT fibrosis. Although fibrosis is a hallmark of obese AT, the role of fibroblasts, cells known to regulate fibrosis in other fibrosis-prone tissues, is not well studied. Here we have developed an in vitro model of AT to study adipocyte-fibroblast crosstalk in a hypoxic environment. Further, this in vitro model was used to investigate the effect of hypoxia on adipocyte mechanical properties via ras homolog gene family member A (RhoA)/Rho-associated coiled-coil kinases (ROCK) signaling pathways. We confirmed that hypoxia creates a diseased phenotype by inhibiting adipocyte maturation and inducing actin stress fiber formation facilitated by myocardin-related transcription factor A (MRTF-A/MKL1) nuclear translocation. This work presents new potential therapeutic targets for obesity by improving adipocyte maturation and limiting mechanical stress in obese AT.
Collapse
|
24
|
The cyclin dependent kinase inhibitor Roscovitine prevents diet-induced metabolic disruption in obese mice. Sci Rep 2021; 11:20365. [PMID: 34645915 PMCID: PMC8514475 DOI: 10.1038/s41598-021-99871-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022] Open
Abstract
Most strategies to treat obesity-related disorders have involved prevention of diet-induced weight gain in lean mice. Treatment of obese individuals will require therapies that reverse the detrimental effects of excess body weight. Cyclin-dependent kinases have been shown to contribute to obesity and its adverse complications. Here, we show that roscovitine; a an orally available cyclin-dependent kinase inhibitor; given to male mice during the last six weeks of a 19-week high fat diet, reduced weight gain and prevented accompanying insulin resistance, hepatic steatosis, visceral adipose tissue (eWAT) inflammation/fibrosis as well as restored insulin secretion and enhanced whole body energy expenditure. Proteomics and phosphoproteomics analysis of eWAT demonstrated that roscovitine suppressed expression of peptides and phosphopeptides linked to inflammation and extracellular matrix proteins. It also identified 17 putative protein kinases perturbed by roscovitine, including CMGC kinases, AGC kinases and CAMK kinases. Pathway enrichment analysis showed that lipid metabolism, TCA cycle, fatty acid beta oxidation and creatine biosynthesis are enriched following roscovitine treatment. For brown adipose tissue (BAT), analysis of upstream kinases controlling the phosphoproteome revealed two major kinase groups, AGC and CMGC kinases. Among the top enriched pathways were insulin signaling, regulation of lipolysis in adipocytes, thyroid hormone signaling, thermogenesis and cAMP-PKG signaling. We conclude that roscovitine is effective at preventing prolonged diet-induced metabolic disruption and restoring mitochondrial activity in BAT and eWAT.
Collapse
|
25
|
Yang Y, Zhang Y, Zhou X, Chen D, Ouyang G, Liu Y, Cui D. Periostin deficiency attenuates lipopolysaccharide- and obesity-induced adipose tissue fibrosis. FEBS Lett 2021; 595:2099-2112. [PMID: 34165806 DOI: 10.1002/1873-3468.14154] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/13/2021] [Accepted: 06/20/2021] [Indexed: 11/09/2022]
Abstract
Periostin (POSTN) is a type of matricellular protein, but its functions in adipose fibrosis remain unclear. Here, we found that POSTN expression is significantly increased in mouse adipose tissue after treatment with lipopolysaccharide (LPS) or a high-fat diet (HFD) and that adipose progenitor cells are the main source of POSTN. In our mouse model of fibrosis, POSTN deletion protected mice from adipose fibrosis, probably through reducing the accumulation of macrophages and promoting adipocyte differentiation of progenitor cells. Taken together, our study demonstrates that POSTN deficiency attenuates adipose tissue fibrosis and improves insulin resistance, providing new insights into the diagnosis and treatment of type II diabetes by targeting adipose tissue fibrosis.
Collapse
Affiliation(s)
- Yuhan Yang
- School of Life Sciences, Xiamen University, China
| | - Yanfei Zhang
- School of Life Sciences, Xiamen University, China
| | | | - Dandan Chen
- School of Life Sciences, Xiamen University, China
| | | | - Yingfu Liu
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, China
| | - Dan Cui
- School of Life Sciences, Xiamen University, China
| |
Collapse
|
26
|
Abstract
Adipose is a key tissue regulating energy homeostasis. In states of obesity, caloric intake exceeds energy expenditure, thereby accelerating lipid accumulation with ongoing extracellular matrix (ECM) remodeling. Excess deposition of lipids and expansion of adipocytes potentially decrease ECM flexibility with local hypoxia and inflammation. Hypoxia and chronic low-grade inflammation accelerate the development of adipose tissue fibrosis and related metabolic dysfunctions. Recent research investigated that some cytokines and proteins are functional in regulating energy homeostasis, meanwhile, are potential targets to fight against adipose tissue fibrosis and insulin resistance. In this review, we focused on the regulatory mechanisms and mediators in remodeling of adipose tissue fibrosis, along with their relevance to clinical manifestations.
Collapse
Affiliation(s)
- Siqi Li
- School of Life Science, Changchun Normal University, Changchun, China
- School of Medical Technology, Beihua University, Jilin, China
- Diagnostic Research Center, Jilin Province People's Hospital, Changchun, China
| | - Hongxia Gao
- School of Medical Technology, Beihua University, Jilin, China
| | - Yutaka Hasegawa
- Department of Internal Medicine, Iwate Medical University, Iwate, Japan
| | - Xiaodan Lu
- School of Life Science, Changchun Normal University, Changchun, China
- School of Medical Technology, Beihua University, Jilin, China
- Diagnostic Research Center, Jilin Province People's Hospital, Changchun, China
| |
Collapse
|
27
|
Zhang L, Li HL, Zhang DD, Cui XC. Therapeutic effects of myocardin-related transcription factor A (MRTF-A) knockout on experimental mice with nonalcoholic steatohepatitis induced by high-fat diet. Hum Exp Toxicol 2021; 40:1634-1645. [PMID: 33779332 DOI: 10.1177/09603271211002886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To explore the effects of myocardin-related transcription factor A (MRTF-A) knockout on mice with nonalcoholic steatohepatitis (NASH) induced by high-fat diet (HFD). METHODS Normal-fat diet (NFD) or HFD was fed to MRTF-A-knockout (MRTF-A-/-) and wild-type (WT) mice for 16 weeks. Liver histopathological status was observed using Hematoxylin and Eosin (HE) staining, Oil Red O staining, Sirius Red staining, and Immunohistochemical staining. The mRNA and protein levels in liver tissues were measured through quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot. RESULTS Compared with WT + HFD group, mice in MRTF-A-/- + HFD group were decreased in body weight, blood glucose, plasma insulin, liver TG and NAFLD activity score (NAS), with liver function recovery. Besides, compared with HFD-fed WT mice, HFD-fed MRTF-A-/- mice were improved in hepatic fibrosis, accompanied by decreased collagen content (%) and down-regulated expressions of α-SMA, COL1A2, TGFβ1, and SMAD3. In mice fed with HFD, the expression of MCP-1, CCR2, F4/80 and CD68 declined in liver tissues of MRTF-A-/- mice as compared with WT mice. Besides, in hepatic macrophages isolated from HFD-fed mice, the observed increased expression of TNF-α, IL-1β, MCP-1, as well as decreased expression of CCR2. Compared with WT + HFD group, MRTF-A-/- + HFD group mice were decreased regarding NF-κB p65 in liver tissues. CONCLUSION MRTF-A knockout reduced macrophage infiltration, down-regulated NF-κB p65 expression, and ameliorated inflammation and fibrosis of liver tissues in mice, thereby becoming a potential therapeutic target for NASH treatment.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Infectious Diseases, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Hua-Long Li
- Department of Infectious Diseases, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Ding-Ding Zhang
- Department of Infectious Diseases, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Xiao-Chun Cui
- Bone and Joint Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| |
Collapse
|
28
|
Liu W, Li D, Cao H, Li H, Wang Y. Expansion and inflammation of white adipose tissue - focusing on adipocyte progenitors. Biol Chem 2020; 402:123-132. [PMID: 33544474 DOI: 10.1515/hsz-2019-0451] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 10/01/2020] [Indexed: 12/22/2022]
Abstract
Adipose tissue is an important organ in our body, participating not only in energy metabolism but also immune regulation. It is broadly classified as white (WAT) and brown (BAT) adipose tissues. WAT is highly heterogeneous, composed of adipocytes, various immune, progenitor and stem cells, as well as the stromal vascular populations. The expansion and inflammation of WAT are hallmarks of obesity and play a causal role in the development of metabolic and cardiovascular diseases. The primary event triggering the inflammatory expansion of WAT remains unclear. The present review focuses on the role of adipocyte progenitors (APS), which give rise to specialized adipocytes, in obesity-associated WAT expansion, inflammation and fibrosis.
Collapse
Affiliation(s)
- Wenjing Liu
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Dahui Li
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Handi Cao
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Haoyun Li
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
29
|
Pyrina I, Chung KJ, Michailidou Z, Koutsilieris M, Chavakis T, Chatzigeorgiou A. Fate of Adipose Progenitor Cells in Obesity-Related Chronic Inflammation. Front Cell Dev Biol 2020; 8:644. [PMID: 32760729 PMCID: PMC7372115 DOI: 10.3389/fcell.2020.00644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/26/2020] [Indexed: 12/16/2022] Open
Abstract
Adipose progenitor cells, or preadipocytes, constitute a small population of immature cells within the adipose tissue. They are a heterogeneous group of cells, in which different subtypes have a varying degree of commitment toward diverse cell fates, contributing to white and beige adipogenesis, fibrosis or maintenance of an immature cell phenotype with proliferation capacity. Mature adipocytes as well as cells of the immune system residing in the adipose tissue can modulate the function and differentiation potential of preadipocytes in a contact- and/or paracrine-dependent manner. In the course of obesity, the accumulation of immune cells within the adipose tissue contributes to the development of a pro-inflammatory microenvironment in the tissue. Under such circumstances, the crosstalk between preadipocytes and immune or parenchymal cells of the adipose tissue may critically regulate the differentiation of preadipocytes into white adipocytes, beige adipocytes, or myofibroblasts, thereby influencing adipose tissue expansion and adipose tissue dysfunction, including downregulation of beige adipogenesis and development of fibrosis. The present review will outline the current knowledge about factors shaping cell fate decisions of adipose progenitor cells in the context of obesity-related inflammation.
Collapse
Affiliation(s)
- Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Zoi Michailidou
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Antonios Chatzigeorgiou
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
30
|
Oguri Y, Shinoda K, Kim H, Alba DL, Bolus WR, Wang Q, Brown Z, Pradhan RN, Tajima K, Yoneshiro T, Ikeda K, Chen Y, Cheang RT, Tsujino K, Kim CR, Greiner VJ, Datta R, Yang CD, Atabai K, McManus MT, Koliwad SK, Spiegelman BM, Kajimura S. CD81 Controls Beige Fat Progenitor Cell Growth and Energy Balance via FAK Signaling. Cell 2020; 182:563-577.e20. [PMID: 32615086 DOI: 10.1016/j.cell.2020.06.021] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/30/2020] [Accepted: 06/09/2020] [Indexed: 01/03/2023]
Abstract
Adipose tissues dynamically remodel their cellular composition in response to external cues by stimulating beige adipocyte biogenesis; however, the developmental origin and pathways regulating this process remain insufficiently understood owing to adipose tissue heterogeneity. Here, we employed single-cell RNA-seq and identified a unique subset of adipocyte progenitor cells (APCs) that possessed the cell-intrinsic plasticity to give rise to beige fat. This beige APC population is proliferative and marked by cell-surface proteins, including PDGFRα, Sca1, and CD81. Notably, CD81 is not only a beige APC marker but also required for de novo beige fat biogenesis following cold exposure. CD81 forms a complex with αV/β1 and αV/β5 integrins and mediates the activation of integrin-FAK signaling in response to irisin. Importantly, CD81 loss causes diet-induced obesity, insulin resistance, and adipose tissue inflammation. These results suggest that CD81 functions as a key sensor of external inputs and controls beige APC proliferation and whole-body energy homeostasis.
Collapse
Affiliation(s)
- Yasuo Oguri
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Beth Israel Deaconess Medical Center, Division of Endocrinology, Diabetes & Metabolism, Harvard Medical School, Boston, MA, USA
| | - Kosaku Shinoda
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, USA
| | - Hyeonwoo Kim
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Diana L Alba
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - W Reid Bolus
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Qiang Wang
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Beth Israel Deaconess Medical Center, Division of Endocrinology, Diabetes & Metabolism, Harvard Medical School, Boston, MA, USA
| | - Zachary Brown
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Rachana N Pradhan
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Kazuki Tajima
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Takeshi Yoneshiro
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Kenji Ikeda
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Molecular Endocrinology and Metabolism, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yong Chen
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rachel T Cheang
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kazuyuki Tsujino
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Osaka, Japan
| | - Caroline R Kim
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Vanille Juliette Greiner
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Ritwik Datta
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher D Yang
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Kamran Atabai
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Michael T McManus
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Suneil K Koliwad
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - Shingo Kajimura
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Beth Israel Deaconess Medical Center, Division of Endocrinology, Diabetes & Metabolism, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Marcelin G, Silveira ALM, Martins LB, Ferreira AV, Clément K. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. J Clin Invest 2020; 129:4032-4040. [PMID: 31498150 DOI: 10.1172/jci129192] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Obesity originates from an imbalance between caloric intake and energy expenditure that promotes adipose tissue expansion, which is necessary to buffer nutrient excess. Patients with higher visceral fat mass are at a higher risk of developing severe complications such as type 2 diabetes and cardiovascular and liver diseases. However, increased fat mass does not fully explain obesity's propensity to promote metabolic diseases. With chronic obesity, adipose tissue undergoes major remodeling, which can ultimately result in unresolved chronic inflammation leading to fibrosis accumulation. These features drive local tissue damage and initiate and/or maintain multiorgan dysfunction. Here, we review the current understanding of adipose tissue remodeling with a focus on obesity-induced adipose tissue fibrosis and its relevance to clinical manifestations.
Collapse
Affiliation(s)
- Geneviève Marcelin
- Nutrition and Obesities: Systemic Approaches (NutriOmics, UMRS U1269), INSERM, Sorbonne Université, Paris, France
| | - Ana Letícia M Silveira
- Nutrition and Obesities: Systemic Approaches (NutriOmics, UMRS U1269), INSERM, Sorbonne Université, Paris, France.,Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laís Bhering Martins
- Nutrition and Obesities: Systemic Approaches (NutriOmics, UMRS U1269), INSERM, Sorbonne Université, Paris, France.,Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adaliene Vm Ferreira
- Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches (NutriOmics, UMRS U1269), INSERM, Sorbonne Université, Paris, France.,Nutrition Department, Hôpital Pitié-Salpêtrière, Assistance Publique Hôpitaux de Paris, Paris, France
| |
Collapse
|
32
|
Discoidin domain receptor 1-deletion ameliorates fibrosis and promotes adipose tissue beiging, brown fat activity, and increased metabolic rate in a mouse model of cardiometabolic disease. Mol Metab 2020; 39:101006. [PMID: 32360427 PMCID: PMC7242876 DOI: 10.1016/j.molmet.2020.101006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Objective Discoidin domain receptor 1 (DDR1) is a collagen binding receptor tyrosine kinase implicated in atherosclerosis, fibrosis, and cancer. Our previous research showed that DDR1 could regulate smooth muscle cell trans-differentiation, fibrosis and calcification in the vascular system in cardiometabolic disease. This spectrum of activity led us to question whether DDR1 might also regulate adipose tissue fibrosis and remodeling. Methods We have used a diet-induced mouse model of cardiometabolic disease to determine whether DDR1 deletion impacts upon adipose tissue remodeling and metabolic dysfunction. Mice were fed a high fat diet (HFD) for 12 weeks, followed by assessment of glucose and insulin tolerance, respiration via indirect calorimetry, and brown fat activity by FDG-PET. Results Feeding HFD induced DDR1 expression in white adipose tissue, which correlated with adipose tissue expansion and fibrosis. Ddr1−/− mice fed an HFD had improved glucose tolerance, reduced body fat, and increased brown fat activity and energy expenditure compared to Ddr1+/+ littermate controls. HFD-fed DDR1−/− mice also had reduced fibrosis, smaller adipocytes with multilocular lipid droplets, and increased UCP-1 expression characteristic of beige fat formation in subcutaneous adipose tissue. In vitro, studying C3H10T1/2 cells stimulated to differentiate, DDR1 inhibition caused a shift from white to beige adipocyte differentiation, whereas DDR1 expression was increased with TGFβ-mediated pro-fibrotic differentiation. Conclusion This study is the first to identify a role for DDR1 as a driver of adipose tissue fibrosis and suppressor of beneficial beige fat formation. DDR1 deletion results in decreased obesity, and increased energy expenditure and brown fat activity. DDR1 expression was increased in adipose and correlated with obesity and fibrosis. DDR1 deletion increased UCP-1 expression in brown and white fat in vivo, and in mesenchymal cells in vitro. Invitro studies suggest that DDR1 suppresses UCP-1 and drives pro-fibrotic differentiation of mesenchymal cells.
Collapse
|
33
|
The Adipocyte Acquires a Fibroblast-Like Transcriptional Signature in Response to a High Fat Diet. Sci Rep 2020; 10:2380. [PMID: 32047213 PMCID: PMC7012923 DOI: 10.1038/s41598-020-59284-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/20/2020] [Indexed: 12/20/2022] Open
Abstract
Visceral white adipose tissue (vWAT) expands and undergoes extensive remodeling during diet-induced obesity. Much is known about the contribution of various stromal vascular cells to the remodeling process, but less is known of the changes that occur within the adipocyte as it becomes progressively dysfunctional. Here, we performed a transcriptome analysis of isolated vWAT adipocytes to assess global pathway changes occurring in response to a chronic high fat diet (HFD). The data demonstrate that the adipocyte responds to the HFD by adopting a fibroblast-like phenotype, characterized by enhanced expression of ECM, focal adhesion and cytoskeletal genes and suppression of many adipocyte programs most notably those associated with mitochondria. This study reveals that during obesity the adipocyte progressively becomes metabolically dysfunctional due to its acquisition of fibrogenic functions. We propose that mechano-responsive transcription factors such as MRTFA and SRF contribute to both upregulation of morphological genes as well as suppression of mitochondrial programs.
Collapse
|
34
|
Wang W, Ishibashi J, Trefely S, Shao M, Cowan AJ, Sakers A, Lim HW, O'Connor S, Doan MT, Cohen P, Baur JA, King MT, Veech RL, Won KJ, Rabinowitz JD, Snyder NW, Gupta RK, Seale P. A PRDM16-Driven Metabolic Signal from Adipocytes Regulates Precursor Cell Fate. Cell Metab 2019; 30:174-189.e5. [PMID: 31155495 PMCID: PMC6836679 DOI: 10.1016/j.cmet.2019.05.005] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 03/28/2019] [Accepted: 05/01/2019] [Indexed: 12/18/2022]
Abstract
The precursor cells for metabolically beneficial beige adipocytes can alternatively become fibrogenic and contribute to adipose fibrosis. We found that cold exposure or β3-adrenergic agonist treatment of mice decreased the fibrogenic profile of precursor cells and stimulated beige adipocyte differentiation. This fibrogenic-to-adipogenic transition was impaired in aged animals, correlating with reduced adipocyte expression of the transcription factor PRDM16. Genetic loss of Prdm16 mimicked the effect of aging in promoting fibrosis, whereas increasing PRDM16 in aged mice decreased fibrosis and restored beige adipose development. PRDM16-expressing adipose cells secreted the metabolite β-hydroxybutyrate (BHB), which blocked precursor fibrogenesis and facilitated beige adipogenesis. BHB catabolism in precursor cells, mediated by BDH1, was required for beige fat differentiation in vivo. Finally, dietary BHB supplementation in aged animals reduced adipose fibrosis and promoted beige fat formation. Together, our results demonstrate that adipocytes secrete a metabolite signal that controls beige fat remodeling.
Collapse
Affiliation(s)
- Wenshan Wang
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeff Ishibashi
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sophie Trefely
- Department of Cancer Biology, University of Pennsylvania Philadelphia, PA, USA; AJ Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
| | - Mengle Shao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexis J Cowan
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ, 08544 USA
| | - Alexander Sakers
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Hee-Woong Lim
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Genetics Department, University of Pennsylvania, Philadelphia, PA, USA
| | - Sean O'Connor
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Mary T Doan
- AJ Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Joseph A Baur
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M Todd King
- Laboratory of Metabolic Control, NIH/NIAAA, Rockville, MD, USA
| | - Richard L Veech
- Laboratory of Metabolic Control, NIH/NIAAA, Rockville, MD, USA
| | - Kyoung-Jae Won
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Genetics Department, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ, 08544 USA
| | | | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Shapira SN, Seale P. Transcriptional Control of Brown and Beige Fat Development and Function. Obesity (Silver Spring) 2019; 27:13-21. [PMID: 30569639 PMCID: PMC6309799 DOI: 10.1002/oby.22334] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022]
Abstract
Adipose tissue, once viewed as an inert organ of energy storage, is now appreciated to be a central node for the dynamic regulation of systemic metabolism. There are three general types of adipose tissue: white, brown, and brown-in-white or "beige" fat. All three types of adipose tissue communicate extensively with other organs in the body, including skin, liver, pancreas, muscle, and brain, to maintain energy homeostasis. When energy intake chronically exceeds energy expenditure, obesity and its comorbidities can develop. Thus, understanding the molecular mechanisms by which different types of adipose tissues develop and function could uncover new therapies for combating disorders of energy imbalance. In this review, the recent findings on the transcriptional and chromatin-mediated regulation of brown and beige adipose tissue activity are highlighted.
Collapse
Affiliation(s)
- Suzanne N. Shapira
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
36
|
Abstract
At the simplest level, obesity is the manifestation of an imbalance between caloric intake and expenditure; however, the pathophysiological mechanisms that govern the development of obesity and associated complications are enormously complex. Fibrosis within the adipose tissue compartment is one such factor that may influence the development of obesity and/or obesity-related comorbidities. Furthermore, the functional consequences of adipose tissue fibrosis are a matter of considerable debate, with evidence that fibrosis serves both adaptive and maladaptive roles. Tissue fibrosis itself is incompletely understood, and multiple cellular and molecular pathways are involved in the development, maintenance, and resolution of the fibrotic state. Within the context of obesity, fibrosis influences molecular and cellular events that relate to adipocytes, inflammatory cells, inflammatory mediators, and supporting adipose stromal tissue. In this Review, we explore what is known about the interplay between the development of adipose tissue fibrosis and obesity, with a view toward future investigative and therapeutic avenues.
Collapse
Affiliation(s)
| | - Michael J Podolsky
- Cardiovascular Research Institute.,Lung Biology Center, and.,Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Kamran Atabai
- Cardiovascular Research Institute.,Lung Biology Center, and.,Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|