1
|
Neugebauer J, Raulien N, Arndt L, Akkermann D, Hobusch C, Lindhorst A, Fröba J, Gericke M. The Impact of Resident Adipose Tissue Macrophages on Adipocyte Homeostasis and Dedifferentiation. Int J Mol Sci 2024; 25:13019. [PMID: 39684730 DOI: 10.3390/ijms252313019] [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: 10/29/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity is concurrent with immunological dysregulation, resulting in chronic low-grade inflammation and cellular dysfunction. In pancreatic islets, this loss of function has been correlated with mature β-cells dedifferentiating into a precursor-like state through constant exposure to inflammatory stressors. As mature adipocytes likewise have the capability to dedifferentiate in vitro and in vivo, we wanted to analyze this cellular change in relation to adipose tissue (AT) inflammation and adipose tissue macrophage (ATM) activity. Using our organotypic AT explant culture method combined with a double-reporter mouse model for labeling ATMs and mature adipocytes, we were able to visualize and quantify dedifferentiated fat (DFAT) cells in AT explants. Preliminary testing showed increased dedifferentiation after tamoxifen (TAM) stimulation, making TAM-dependent lineage-tracing models unsuitable for quantification of naturally occurring DFAT cells. The regulatory role of ATMs in adipocyte dedifferentiation was shown through macrophage depletion using Plexxicon 5622 or clodronate liposomes, which significantly increased DFAT cell levels. Subsequent bulk RNA sequencing of macrophage-depleted explants revealed enrichment of the tumor necrosis factor α (TNFα) signaling pathway as well as downregulation of associated genes. Direct stimulation with TNFα decreased adipocyte dedifferentiation, while application of a TNFα-neutralizing antibody did not significantly alter DFAT cell levels. Our findings suggest a regulatory role of resident ATMs in maintaining the mature adipocyte phenotype and preventing excessive adipocyte dedifferentiation. The specific regulatory pathways as well as the impact that DFAT cells might have on ATMs, and vice versa, are subject to further investigation.
Collapse
Affiliation(s)
- Julia Neugebauer
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Nora Raulien
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Lilli Arndt
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Dagmar Akkermann
- Paul-Flechsig-Institute, Leipzig University, 04103 Leipzig, Germany
| | | | | | - Janine Fröba
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
2
|
Plissonneau C, Santosa S. Regional primary preadipocyte characteristics in humans with obesity and type 2 diabetes mellitus. Heliyon 2024; 10:e39710. [PMID: 39553621 PMCID: PMC11564010 DOI: 10.1016/j.heliyon.2024.e39710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
The excessive accumulation of adipose tissue in obesity appears to result in adipose tissue dysfunction perpetuating the onset of obesity-related diseases, including type 2 diabetes (T2DM). In humans, adipose tissue is stored in several depots including subcutaneous and visceral. These depots contribute to the pathology of obesity differently owing to differences in the tissue microenvironment, a main one being preadipocyte function. In examining adipocyte and preadipocyte characteristics, many have used the 3T3-L1 murine cell lines. Though these cell lines provide valuable mechanistic data, the results remain to be translated to humans. Experiments using primary human preadipocytes has shown that obesity and T2DM impact preadipocyte phenotypes. The objective of this review is to describe the differences in regional characteristics of primary preadipocytes collected from humans with obesity and to discuss how these characteristics might be affected in type 2 diabetes mellitus. In doing so, we will show that the characteristics of regional primary preadipocytes in humans are differentially affected by obesity and the development of T2DM.
Collapse
Affiliation(s)
- Claire Plissonneau
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Montreal, Quebec, Canada
- Metabolism, Obesity, and Nutrition Lab, School of Health, Concordia University, Montreal, Quebec, Canada
- Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| | - Sylvia Santosa
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Montreal, Quebec, Canada
- Metabolism, Obesity, and Nutrition Lab, School of Health, Concordia University, Montreal, Quebec, Canada
- Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| |
Collapse
|
3
|
Huang P, Zhu Y, Qin J. Research advances in understanding crosstalk between organs and pancreatic β-cell dysfunction. Diabetes Obes Metab 2024; 26:4147-4164. [PMID: 39044309 DOI: 10.1111/dom.15787] [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: 06/18/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024]
Abstract
Obesity has increased dramatically worldwide. Being overweight or obese can lead to various conditions, including dyslipidaemia, hypertension, glucose intolerance and metabolic syndrome (MetS), which may further lead to type 2 diabetes mellitus (T2DM). Previous studies have identified a link between β-cell dysfunction and the severity of MetS, with multiple organs and tissues affected. Identifying the associations between pancreatic β-cell dysfunction and organs is critical. Research has focused on the interaction between the liver, gut and pancreatic β-cells. However, the mechanisms and related core targets are still not perfectly elucidated. The aims of this review were to summarize the mechanisms of β-cell dysfunction and to explore the potential pathogenic pathways and targets that connect the liver, gut, adipose tissue, muscle, and brain to pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yunling Zhu
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jian Qin
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
4
|
Yang Z, Chen F, Zhang Y, Ou M, Tan P, Xu X, Li Q, Zhou S. Therapeutic targeting of white adipose tissue metabolic dysfunction in obesity: mechanisms and opportunities. MedComm (Beijing) 2024; 5:e560. [PMID: 38812572 PMCID: PMC11134193 DOI: 10.1002/mco2.560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 04/09/2024] [Accepted: 04/14/2024] [Indexed: 05/31/2024] Open
Abstract
White adipose tissue is not only a highly heterogeneous organ containing various cells, such as adipocytes, adipose stem and progenitor cells, and immune cells, but also an endocrine organ that is highly important for regulating metabolic and immune homeostasis. In individuals with obesity, dynamic cellular changes in adipose tissue result in phenotypic switching and adipose tissue dysfunction, including pathological expansion, WAT fibrosis, immune cell infiltration, endoplasmic reticulum stress, and ectopic lipid accumulation, ultimately leading to chronic low-grade inflammation and insulin resistance. Recently, many distinct subpopulations of adipose tissue have been identified, providing new insights into the potential mechanisms of adipose dysfunction in individuals with obesity. Therefore, targeting white adipose tissue as a therapeutic agent for treating obesity and obesity-related metabolic diseases is of great scientific interest. Here, we provide an overview of white adipose tissue remodeling in individuals with obesity including cellular changes and discuss the underlying regulatory mechanisms of white adipose tissue metabolic dysfunction. Currently, various studies have uncovered promising targets and strategies for obesity treatment. We also outline the potential therapeutic signaling pathways of targeting adipose tissue and summarize existing therapeutic strategies for antiobesity treatment including pharmacological approaches, lifestyle interventions, and novel therapies.
Collapse
Affiliation(s)
- Zi‐Han Yang
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang‐Zhou Chen
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi‐Xiang Zhang
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Min‐Yi Ou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Poh‐Ching Tan
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue‐Wen Xu
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Qing‐Feng Li
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuang‐Bai Zhou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
5
|
Ispas S, Tuta LA, Botnarciuc M, Ispas V, Staicovici S, Ali S, Nelson-Twakor A, Cojocaru C, Herlo A, Petcu A. Metabolic Disorders, the Microbiome as an Endocrine Organ, and Their Relations with Obesity: A Literature Review. J Pers Med 2023; 13:1602. [PMID: 38003917 PMCID: PMC10672252 DOI: 10.3390/jpm13111602] [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: 10/12/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
The etiology of metabolic disorders, such as obesity, has been predominantly associated with the gut microbiota, which is acknowledged as an endocrine organ that plays a crucial role in modulating energy homeostasis and host immune responses. The presence of dysbiosis has the potential to impact the functioning of the intestinal barrier and the gut-associated lymphoid tissues by allowing the transit of bacterial structural components, such as lipopolysaccharides. This, in turn, may trigger inflammatory pathways and potentially lead to the onset of insulin resistance. Moreover, intestinal dysbiosis has the potential to modify the production of gastrointestinal peptides that are linked to the feeling of fullness, hence potentially leading to an increase in food consumption. In this literature review, we discuss current developments, such as the impact of the microbiota on lipid metabolism as well as the processes by which its changes led to the development of metabolic disorders. Several methods have been developed that could be used to modify the gut microbiota and undo metabolic abnormalities. METHODS After researching different databases, we examined the PubMed collection of articles and conducted a literature review. RESULTS After applying our exclusion and inclusion criteria, the initial search yielded 1345 articles. We further used various filters to narrow down our titles analysis and, to be specific to our study, selected the final ten studies, the results of which are included in the Results section. CONCLUSIONS Through gut barrier integrity, insulin resistance, and other influencing factors, the gut microbiota impacts the host's metabolism and obesity. Although the area of the gut microbiota and its relationship to obesity is still in its initial stages of research, it offers great promise for developing new therapeutic targets that may help prevent and cure obesity by restoring the gut microbiota to a healthy condition.
Collapse
Affiliation(s)
- Sorina Ispas
- Department of Anatomy, Faculty of General Medicine, “Ovidius” University, 900470 Constanta, Romania; (S.I.); (V.I.)
| | - Liliana Ana Tuta
- Department of Clinical Medicine, Faculty of General Medicine, “Ovidius” University, 900470 Constanta, Romania
- Head of Nephrology Section, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Mihaela Botnarciuc
- Department of Microbiology, Faculty of General Medicine, “Ovidius” University, 900470 Constanta, Romania;
- Head of Blood Transfusions Section, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Viorel Ispas
- Department of Anatomy, Faculty of General Medicine, “Ovidius” University, 900470 Constanta, Romania; (S.I.); (V.I.)
- Vascular Surgery Department, Cai Ferate Hospital, 35–37 I. C. Bratianu Boulevard, 900270 Constanta, Romania
| | - Sorana Staicovici
- Family Medicine, “Regina Maria” Polyclinic, 900189 Constanta, Romania;
- Department of Histology, Faculty of General Medicine, “Ovidius” University, 900470 Constanta, Romania
| | - Sevigean Ali
- Preclinics Department II, Faculty of General Medicine, “Ovidius” University, 900470 Constanta, Romania;
- County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | | | | | - Alexandra Herlo
- Department XIII, Discipline of Infectious Diseases, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Adina Petcu
- Department of Mathematics, Biostatistics and Medical Informatics, Faculty of Pharmacy, “Ovidius” University, 900470 Constanta, Romania;
| |
Collapse
|
6
|
Li X, Ren Y, Chang K, Wu W, Griffiths HR, Lu S, Gao D. Adipose tissue macrophages as potential targets for obesity and metabolic diseases. Front Immunol 2023; 14:1153915. [PMID: 37153549 PMCID: PMC10154623 DOI: 10.3389/fimmu.2023.1153915] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
Macrophage infiltration into adipose tissue is a key pathological factor inducing adipose tissue dysfunction and contributing to obesity-induced inflammation and metabolic disorders. In this review, we aim to present the most recent research on macrophage heterogeneity in adipose tissue, with a focus on the molecular targets applied to macrophages as potential therapeutics for metabolic diseases. We begin by discussing the recruitment of macrophages and their roles in adipose tissue. While resident adipose tissue macrophages display an anti-inflammatory phenotype and promote the development of metabolically favorable beige adipose tissue, an increase in pro-inflammatory macrophages in adipose tissue has negative effects on adipose tissue function, including inhibition of adipogenesis, promotion of inflammation, insulin resistance, and fibrosis. Then, we presented the identities of the newly discovered adipose tissue macrophage subtypes (e.g. metabolically activated macrophages, CD9+ macrophages, lipid-associated macrophages, DARC+ macrophages, and MFehi macrophages), the majority of which are located in crown-like structures within adipose tissue during obesity. Finally, we discussed macrophage-targeting strategies to ameliorate obesity-related inflammation and metabolic abnormalities, with a focus on transcriptional factors such as PPARγ, KLF4, NFATc3, and HoxA5, which promote macrophage anti-inflammatory M2 polarization, as well as TLR4/NF-κB-mediated inflammatory pathways that activate pro-inflammatory M1 macrophages. In addition, a number of intracellular metabolic pathways closely associated with glucose metabolism, oxidative stress, nutrient sensing, and circadian clock regulation were examined. Understanding the complexities of macrophage plasticity and functionality may open up new avenues for the development of macrophage-based treatments for obesity and other metabolic diseases.
Collapse
Affiliation(s)
- Xirong Li
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yakun Ren
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Kewei Chang
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
| | - Wenlong Wu
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Helen R. Griffiths
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Shemin Lu
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Dan Gao
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
| |
Collapse
|
7
|
Zhang YX, Ou MY, Yang ZH, Sun Y, Li QF, Zhou SB. Adipose tissue aging is regulated by an altered immune system. Front Immunol 2023; 14:1125395. [PMID: 36875140 PMCID: PMC9981968 DOI: 10.3389/fimmu.2023.1125395] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/30/2023] [Indexed: 02/19/2023] Open
Abstract
Adipose tissue is a widely distributed organ that plays a critical role in age-related physiological dysfunctions as an important source of chronic sterile low-grade inflammation. Adipose tissue undergoes diverse changes during aging, including fat depot redistribution, brown and beige fat decrease, functional decline of adipose progenitor and stem cells, senescent cell accumulation, and immune cell dysregulation. Specifically, inflammaging is common in aged adipose tissue. Adipose tissue inflammaging reduces adipose plasticity and pathologically contributes to adipocyte hypertrophy, fibrosis, and ultimately, adipose tissue dysfunction. Adipose tissue inflammaging also contributes to age-related diseases, such as diabetes, cardiovascular disease and cancer. There is an increased infiltration of immune cells into adipose tissue, and these infiltrating immune cells secrete proinflammatory cytokines and chemokines. Several important molecular and signaling pathways mediate the process, including JAK/STAT, NFκB and JNK, etc. The roles of immune cells in aging adipose tissue are complex, and the underlying mechanisms remain largely unclear. In this review, we summarize the consequences and causes of inflammaging in adipose tissue. We further outline the cellular/molecular mechanisms of adipose tissue inflammaging and propose potential therapeutic targets to alleviate age-related problems.
Collapse
Affiliation(s)
- Yi-Xiang Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min-Yi Ou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zi-Han Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Sun
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qing-Feng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuang-Bai Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Emerging Roles of Adipose Tissue in the Pathogenesis of Psoriasis and Atopic Dermatitis in Obesity. JID INNOVATIONS 2022; 2:100064. [PMID: 35024685 PMCID: PMC8659781 DOI: 10.1016/j.xjidi.2021.100064] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 12/29/2022] Open
Abstract
Obesity is a growing epidemic worldwide, and it is also considered a major environmental factor contributing to the pathogenesis of inflammatory skin diseases, including psoriasis (PSO) and atopic dermatitis (AD). Moreover, obesity worsens the course and impairs the treatment response of these inflammatory skin diseases. Emerging evidence highlights that hypertrophied adipocytes and infiltrated immune cells secrete a variety of molecules, including fatty acids and adipokines, such as leptin, adiponectin, and a panel of cytokines/chemokines that modulate our immune system. In this review, we describe how adipose hypertrophy leads to a chronic low-grade inflammatory state in obesity and how obesity-related inflammatory factors are involved in the pathogenesis of PSO and/or AD. Finally, we discuss the potential role of antimicrobial peptides, mechanical stress and impairment of epidermal barrier function mediated by fast expansion, and dermal fat in modulating skin inflammation. Together, this review summarizes the current literature on how obesity is associated with the pathogenesis of PSO and AD, highlighting the potentially important but overlooked immunomodulatory role of adipose tissue in the skin.
Collapse
Key Words
- AD, atopic dermatitis
- AMP, antimicrobial peptide
- AT, adipose tissue
- BAT, brown adipose tissue
- BMI, body mass index
- CI, confidence interval
- DC, dendritic cell
- DIO, diet-induced obesity
- FFA, free fatty acid
- HFD, high-fat diet
- KC, keratinocyte
- OA, oleic acid
- PA, palmitic acid
- PSO, psoriasis
- SCORAD, SCORing Atopic Dermatitis
- TC, total cholesterol
- TEWL, transepidermal water loss
- TG, triglyceride
- TLR, toll-like receptor
- Th, T helper
- WAT, white adipose tissue
- dFB, dermal fibroblast
- dWAT, dermal white adipose tissue
- sWAT, subcutaneous white adipose tissue
Collapse
|
9
|
Yamada T. Intramuscular adipogenesis in cattle: Effects of body fat distribution and macrophage infiltration. Anim Sci J 2022; 93:e13785. [PMID: 36443236 DOI: 10.1111/asj.13785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/18/2022] [Accepted: 11/01/2022] [Indexed: 11/30/2022]
Abstract
Ectopic fat is defined by the deposition of adipose tissue within non-adipose tissue such as skeletal muscle. Japanese Black cattle (Wagyu) are characterized by the ability to accumulate high amounts of intramuscular adipose tissue. Obese conditions enhance the accumulation of ectopic fat. This review shows the effects of subcutaneous and visceral fat distribution on Wagyu intramuscular adipogenesis. Obese conditions also stimulate the macrophage infiltration into adipose tissues. Adipose tissue macrophages have reported to regulate adipose tissue growth and ectopic fat accumulation in humans and rodents. Wagyu is characterized by the higher capacity for intramuscular adipogenesis than Holsteins. This review discusses the depot-specific effects of macrophage infiltration among subcutaneous, visceral, and intramuscular adipose tissue on intramuscular adipogenesis in Wagyu and Holstein cattle. Recently, metabolome analysis has been used to identify obesity-related biomarkers by comparing the biological samples between lean and obese patients. This review introduces the metabolomic profiles of plasma and intramuscular adipose tissue between Wagyu and Holsteins.
Collapse
Affiliation(s)
- Tomoya Yamada
- National Agriculture and Food Research Organization Oda Shimane Japan
| |
Collapse
|
10
|
Lizcano F. Roles of estrogens, estrogen-like compounds, and endocrine disruptors in adipocytes. Front Endocrinol (Lausanne) 2022; 13:921504. [PMID: 36213285 PMCID: PMC9533025 DOI: 10.3389/fendo.2022.921504] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Women are subject to constitutional changes after menopause, which increases conditions and diseases prone to cardiovascular risks such as obesity and diabetes mellitus. Both estrogens and androgens influence the individual's metabolic mechanism, which controls the fat distribution and the hypothalamic organization of the regulatory centers of hunger and satiety. While androgens tend to accumulate fat in the splanchnic and the visceral region with an increase in cardiovascular risk, estrogens generate more subcutaneous and extremity distribution of adipose tissue. The absence of estrogen during menopause seems to be the main factor that gives rise to the greater predisposition of women to suffer cardiovascular alterations. However, the mechanisms by which estrogens regulate the energy condition of people are not recognized. Estrogens have several mechanisms of action, which mainly include the modification of specific receptors that belong to the steroid receptor superfamily. The alpha estrogen receptors (ERα) and the beta receptors (ERβ) have a fundamental role in the metabolic control of the individual, with a very characteristic corporal distribution that exerts an influence on the metabolism of lipids and glucose. Despite the significant amount of knowledge in this field, many of the regulatory mechanisms exerted by estrogens and ER continue to be clarified. This review will discuss the role of estrogens and their receptors on the central regulation of caloric expenditure and the influence they exert on the differentiation and function of adipocytes. Furthermore, chemical substances with a hormonal activity that cause endocrine disruption with affectation on estrogen receptors will be considered. Finally, the different medical therapies for the vasomotor manifestations of menopause and their role in reducing obesity, diabetes, and cardiovascular risk will be analyzed.
Collapse
|
11
|
Altun I, Yan X, Ussar S. Immune Cell Regulation of White Adipose Progenitor Cell Fate. Front Endocrinol (Lausanne) 2022; 13:859044. [PMID: 35422761 PMCID: PMC9001836 DOI: 10.3389/fendo.2022.859044] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/28/2022] [Indexed: 02/03/2023] Open
Abstract
Adipose tissue is essential for energy storage and endocrine regulation of metabolism. Imbalance in energy intake and expenditure result in obesity causing adipose tissue dysfunction. This alters cellular composition of the stromal cell populations and their function. Moreover, the individual cellular composition of each adipose tissue depot, regulated by environmental factors and genetics, determines the ability of the depots to expand and maintain its endocrine and storage function. Thus, stromal cells modulate adipocyte function and vice versa. In this mini-review we discuss heterogeneity in terms of composition and fate of adipose progenitor subtypes and their interactions with and regulation by different immune cell populations. Immune cells are the most diverse cell populations in adipose tissue and play essential roles in regulating adipose tissue function via interaction with adipocytes but also with adipocyte progenitors. We specifically discuss the role of macrophages, mast cells, innate lymphoid cells and T cells in the regulation of adipocyte progenitor proliferation, differentiation and lineage commitment. Understanding the factors and cellular interactions regulating preadipocyte expansion and fate decision will allow the identification of novel mechanisms and therapeutic strategies to promote healthy adipose tissue expansion without systemic metabolic impairment.
Collapse
Affiliation(s)
- Irem Altun
- Research Group Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Xiaocheng Yan
- Research Group Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Siegfried Ussar
- Research Group Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Medicine, Technische Universität München, Munich, Germany
- *Correspondence: Siegfried Ussar,
| |
Collapse
|
12
|
Cataldi S, Aprile M, Melillo D, Mucel I, Giorgetti-Peraldi S, Cormont M, Italiani P, Blüher M, Tanti JF, Ciccodicola A, Costa V. TNFα Mediates Inflammation-Induced Effects on PPARG Splicing in Adipose Tissue and Mesenchymal Precursor Cells. Cells 2021; 11:cells11010042. [PMID: 35011604 PMCID: PMC8750445 DOI: 10.3390/cells11010042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 01/18/2023] Open
Abstract
Low-grade chronic inflammation and reduced differentiation capacity are hallmarks of hypertrophic adipose tissue (AT) and key contributors of insulin resistance. We identified PPARGΔ5 as a dominant-negative splicing isoform overexpressed in the AT of obese/diabetic patients able to impair adipocyte differentiation and PPARγ activity in hypertrophic adipocytes. Herein, we investigate the impact of macrophage-secreted pro-inflammatory factors on PPARG splicing, focusing on PPARGΔ5. We report that the epididymal AT of LPS-treated mice displays increased PpargΔ5/cPparg ratio and reduced expression of Pparg-regulated genes. Interestingly, pro-inflammatory factors secreted from murine and human pro-inflammatory macrophages enhance the PPARGΔ5/cPPARG ratio in exposed adipogenic precursors. TNFα is identified herein as factor able to alter PPARG splicing—increasing PPARGΔ5/cPPARG ratio—through PI3K/Akt signaling and SRp40 splicing factor. In line with in vitro data, TNFA expression is higher in the SAT of obese (vs. lean) patients and positively correlates with PPARGΔ5 levels. In conclusion, our results indicate that inflammatory factors secreted by metabolically-activated macrophages are potent stimuli that modulate the expression and splicing of PPARG. The resulting imbalance between canonical and dominant negative isoforms may crucially contribute to impair PPARγ activity in hypertrophic AT, exacerbating the defective adipogenic capacity of precursor cells.
Collapse
Affiliation(s)
- Simona Cataldi
- Institute of Genetics and Biophysics ‘‘Adriano Buzzati-Traverso’’, CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (M.A.); (A.C.)
| | - Marianna Aprile
- Institute of Genetics and Biophysics ‘‘Adriano Buzzati-Traverso’’, CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (M.A.); (A.C.)
| | - Daniela Melillo
- Institute of Biochemistry and Cell Biology, CNR, Via P. Castellino 111, 80131 Naples, Italy; (D.M.); (P.I.)
| | - Inès Mucel
- Université Côte d’Azur, Inserm UMR1065, C3M, Team Cellular and Molecular Pathophysiology of Obesity, 06204 Nice, France; (I.M.); (S.G.-P.); (M.C.); (J.-F.T.)
| | - Sophie Giorgetti-Peraldi
- Université Côte d’Azur, Inserm UMR1065, C3M, Team Cellular and Molecular Pathophysiology of Obesity, 06204 Nice, France; (I.M.); (S.G.-P.); (M.C.); (J.-F.T.)
| | - Mireille Cormont
- Université Côte d’Azur, Inserm UMR1065, C3M, Team Cellular and Molecular Pathophysiology of Obesity, 06204 Nice, France; (I.M.); (S.G.-P.); (M.C.); (J.-F.T.)
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology, CNR, Via P. Castellino 111, 80131 Naples, Italy; (D.M.); (P.I.)
| | - Matthias Blüher
- Medical Department III-Endocrinology, Nephrology and Rheumatology, University of Leipzig, 04103 Leipzig, Germany;
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Philipp-Rosenthal-Str. 27, 04103 Leipzig, Germany
| | - Jean-François Tanti
- Université Côte d’Azur, Inserm UMR1065, C3M, Team Cellular and Molecular Pathophysiology of Obesity, 06204 Nice, France; (I.M.); (S.G.-P.); (M.C.); (J.-F.T.)
| | - Alfredo Ciccodicola
- Institute of Genetics and Biophysics ‘‘Adriano Buzzati-Traverso’’, CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (M.A.); (A.C.)
- Department of Science and Technology, University of Naples ‘‘Parthenope’’, 80143 Naples, Italy
| | - Valerio Costa
- Institute of Genetics and Biophysics ‘‘Adriano Buzzati-Traverso’’, CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (M.A.); (A.C.)
- Correspondence: ; Tel.: +39-0816132617
| |
Collapse
|
13
|
McLaughlin T, Schnittger I, Nagy A, Zanley E, Xu Y, Song Y, Nieman K, Tremmel JA, Dey D, Boyd J, Sacks H. Relationship Between Coronary Atheroma, Epicardial Adipose Tissue Inflammation, and Adipocyte Differentiation Across the Human Myocardial Bridge. J Am Heart Assoc 2021; 10:e021003. [PMID: 34726081 PMCID: PMC8751937 DOI: 10.1161/jaha.121.021003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Background Inflammation in epicardial adipose tissue (EAT) may contribute to coronary atherosclerosis. Myocardial bridge is a congenital anomaly in which the left anterior descending coronary artery takes a "tunneled" course under a bridge of myocardium: while atherosclerosis develops in the proximal left anterior descending coronary artery, the bridged portion is spared, highlighting the possibility that geographic separation from inflamed EAT is protective. We tested the hypothesis that inflammation in EAT was related to atherosclerosis by comparing EAT from proximal and bridge depots in individuals with myocardial bridge and varying degrees of atherosclerotic plaque. Methods and Results Maximal plaque burden was quantified by intravascular ultrasound, and inflammation was quantified by pericoronary EAT signal attenuation (pericoronary adipose tissue attenuation) from cardiac computed tomography scans. EAT overlying the proximal left anterior descending coronary artery and myocardial bridge was harvested for measurement of mRNA and microRNA (miRNA) using custom chips by Nanostring; inflammatory cytokines were measured in tissue culture supernatants. Pericoronary adipose tissue attenuation was increased, indicating inflammation, in proximal versus bridge EAT, in proportion to atherosclerotic plaque. Individuals with moderate-high versus low plaque burden exhibited greater expression of inflammation and hypoxia genes, and lower expression of adipogenesis genes. Comparison of gene expression in proximal versus bridge depots revealed differences only in participants with moderate-high plaque: inflammation was higher in proximal and adipogenesis lower in bridge EAT. Secreted inflammatory cytokines tended to be higher in proximal EAT. Hypoxia-inducible factor 1a was highly associated with inflammatory gene expression. Seven miRNAs were differentially expressed by depot: 3192-5P, 518D-3P, and 532-5P were upregulated in proximal EAT, whereas miR 630, 575, 16-5P, and 320E were upregulated in bridge EAT. miR 630 correlated directly with plaque burden and inversely with adipogenesis genes. miR 3192-5P, 518D-3P, and 532-5P correlated inversely with hypoxia/oxidative stress, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PCG1a), adipogenesis, and angiogenesis genes. Conclusions Inflammation is specifically elevated in EAT overlying atherosclerotic plaque, suggesting that EAT inflammation is caused by atherogenic molecular signals, including hypoxia-inducible factor 1a and/or miRNAs in an "inside-to-out" relationship. Adipogenesis was suppressed in the bridge EAT, but only in the presence of atherosclerotic plaque, supporting cross talk between the vasculature and EAT. miR 630 in EAT, expressed differentially according to burden of atherosclerotic plaque, and 3 other miRNAs appear to inhibit key genes related to adipogenesis, angiogenesis, hypoxia/oxidative stress, and thermogenesis in EAT, highlighting a role for miRNA in mediating cross talk between the coronary vasculature and EAT.
Collapse
Affiliation(s)
- Tracey McLaughlin
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Ingela Schnittger
- Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA
| | - Anna Nagy
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Elizabeth Zanley
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Yue Xu
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Yanqiu Song
- Cardiovascular Institute Tianjin Chest Hospital Tianjin China
| | - Koen Nieman
- Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA
| | - Jennifer A Tremmel
- Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA
| | - Damini Dey
- Department of Biomedical Sciences and Medicine Cedars-Sinai Medical Center Biomedical Imaging Research Institute Los Angeles CA
| | - Jack Boyd
- Department of Cardiothoracic Surgery Stanford University School of Medicine Stanford CA
| | - Harold Sacks
- Division of Endocrinology Department of Medicine David Geffen School of Medicine at UCLA Los Angeles CA
| |
Collapse
|
14
|
Chen Q, Liu S, Cao L, Yu M, Wang H. Effects of macrophage regulation on fat grafting survival: Improvement, mechanisms, and potential application-A review. J Cosmet Dermatol 2021; 21:54-61. [PMID: 34129721 DOI: 10.1111/jocd.14295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 06/08/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Autologous fat grafting has become a popular tool in plastic surgery to solve soft tissue defects and achieve skin rejuvenation, but the volume loss after transplantation remains a disturbing problem. In recent years, some new strategies have improved the outcome to some extent, but the fat graft retention is still far from ideal, so there remains a wide development prospect in this field. Macrophages are closely related to the local microenvironment and tissue regeneration, and their role in fat grafting has been increasingly highlighted. AIMS This article was aimed to review the efficacy, possible mechanisms, and potential application of macrophage regulation on fat grafting, as well as concerns and future perspectives of this filed. METHODS A retrospective review of the published data was conducted. RESULTS Most studies indicated that up-regulating M2 macrophages during fat grafting would improve fat retention via promoting neovascularization. M2 macrophages could secrete several pro-angiogenic factors, accelerate extracellular matrix (ECM) remodeling, and directly function on endothelial cells to encourage vascular expansion. In addition, macrophages could influence the proliferation, apoptosis, and adipogenic differentiation of preadipocytes. CONCLUSIONS During autologous fat grafting, appropriately regulating macrophages may become a promising method to increase fat retention. Nevertheless, the M2 macrophage polarizing agents, treatment opportunity, and contraindications require further discussion. We hope our work could promote more in-depth studies in this field, and we are looking forward to a standard procedure for the macrophage therapy in clinical practice.
Collapse
Affiliation(s)
- Qiuyu Chen
- State Key Laboratory of Oral Diseases, Department of Cosmetic and Plastic Surgery, Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shuo Liu
- State Key Laboratory of Oral Diseases, Department of Cosmetic and Plastic Surgery, Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lideng Cao
- State Key Laboratory of Oral Diseases, Department of Cosmetic and Plastic Surgery, Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mei Yu
- State Key Laboratory of Oral Diseases, Department of Cosmetic and Plastic Surgery, Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hang Wang
- State Key Laboratory of Oral Diseases, Department of Cosmetic and Plastic Surgery, Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Maseroli E, Comeglio P, Corno C, Cellai I, Filippi S, Mello T, Galli A, Rapizzi E, Presenti L, Truglia MC, Lotti F, Facchiano E, Beltrame B, Lucchese M, Saad F, Rastrelli G, Maggi M, Vignozzi L. Testosterone treatment is associated with reduced adipose tissue dysfunction and nonalcoholic fatty liver disease in obese hypogonadal men. J Endocrinol Invest 2021; 44:819-842. [PMID: 32772323 PMCID: PMC7946690 DOI: 10.1007/s40618-020-01381-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/31/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE In both preclinical and clinical settings, testosterone treatment (TTh) of hypogonadism has shown beneficial effects on insulin sensitivity and visceral and liver fat accumulation. This prospective, observational study was aimed at assessing the change in markers of fat and liver functioning in obese men scheduled for bariatric surgery. METHODS Hypogonadal patients with consistent symptoms (n = 15) undergoing 27.63 ± 3.64 weeks of TTh were compared to untreated eugonadal (n = 17) or asymptomatic hypogonadal (n = 46) men. A cross-sectional analysis among the different groups was also performed, especially for data derived from liver and fat biopsies. Preadipocytes isolated from adipose tissue biopsies were used to evaluate insulin sensitivity, adipogenic potential and mitochondrial function. NAFLD was evaluated by triglyceride assay and by calculating NAFLD activity score in liver biopsies. RESULTS In TTh-hypogonadal men, histopathological NAFLD activity and steatosis scores, as well as liver triglyceride content were lower than in untreated-hypogonadal men and comparable to eugonadal ones. TTh was also associated with a favorable hepatic expression of lipid handling-related genes. In visceral adipose tissue and preadipocytes, TTh was associated with an increased expression of lipid catabolism and mitochondrial bio-functionality markers. Preadipocytes from TTh men also exhibited a healthier morpho-functional phenotype of mitochondria and higher insulin-sensitivity compared to untreated-hypogonadal ones. CONCLUSIONS The present data suggest that TTh in severely obese, hypogonadal individuals induces metabolically healthier preadipocytes, improving insulin sensitivity, mitochondrial functioning and lipid handling. A potentially protective role for testosterone on the progression of NAFLD, improving hepatic steatosis and reducing intrahepatic triglyceride content, was also envisaged. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02248467, September 25th 2014.
Collapse
Affiliation(s)
- E Maseroli
- Andrology, Women's Endocrinology and Gender Incongruence Unit, Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50134, Florence, Italy
| | - P Comeglio
- Andrology, Women's Endocrinology and Gender Incongruence Unit, Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50134, Florence, Italy
| | - C Corno
- Andrology, Women's Endocrinology and Gender Incongruence Unit, Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50134, Florence, Italy
| | - I Cellai
- Andrology, Women's Endocrinology and Gender Incongruence Unit, Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50134, Florence, Italy
| | - S Filippi
- Interdepartmental Laboratory of Functional and Cellular Pharmacology of Reproduction, University of Florence, Viale Pieraccini 6, 50134, Florence, Italy
| | - T Mello
- Gastroenterology Unit, Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50134, Florence, Italy
| | - A Galli
- Gastroenterology Unit, Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50134, Florence, Italy
| | - E Rapizzi
- Endocrinology Unit, Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50134, Florence, Italy
| | - L Presenti
- General, Bariatric and Metabolic Surgery Unit, Santa Maria Nuova Hospital, , Piazza Santa Maria Nuova, 1, 50122, Florence, Italy
| | - M C Truglia
- General, Bariatric and Metabolic Surgery Unit, Santa Maria Nuova Hospital, , Piazza Santa Maria Nuova, 1, 50122, Florence, Italy
| | - F Lotti
- Andrology, Women's Endocrinology and Gender Incongruence Unit, Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50134, Florence, Italy
| | - E Facchiano
- General, Bariatric and Metabolic Surgery Unit, Santa Maria Nuova Hospital, , Piazza Santa Maria Nuova, 1, 50122, Florence, Italy
| | - B Beltrame
- General, Bariatric and Metabolic Surgery Unit, Santa Maria Nuova Hospital, , Piazza Santa Maria Nuova, 1, 50122, Florence, Italy
| | - M Lucchese
- General, Bariatric and Metabolic Surgery Unit, Santa Maria Nuova Hospital, , Piazza Santa Maria Nuova, 1, 50122, Florence, Italy
| | - F Saad
- Medical Affairs, Bayer AG, Kaiser-Wilhelm-Allee 1, 51373, Leverkusen, Germany
| | - G Rastrelli
- Andrology, Women's Endocrinology and Gender Incongruence Unit, Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50134, Florence, Italy
| | - M Maggi
- Endocrinology Unit, Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50134, Florence, Italy
- I.N.B.B. (Istituto Nazionale Biostrutture E Biosistemi), Viale delle Medaglie d'Oro 305, 00136, Rome, Italy
| | - L Vignozzi
- Andrology, Women's Endocrinology and Gender Incongruence Unit, Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50134, Florence, Italy.
- I.N.B.B. (Istituto Nazionale Biostrutture E Biosistemi), Viale delle Medaglie d'Oro 305, 00136, Rome, Italy.
| |
Collapse
|
16
|
Barroso Oquendo M, Siegel-Axel D, Gerst F, Lorza-Gil E, Moller A, Wagner R, Machann J, Fend F, Königsrainer A, Heni M, Häring HU, Ullrich S, Birkenfeld AL. Pancreatic fat cells of humans with type 2 diabetes display reduced adipogenic and lipolytic activity. Am J Physiol Cell Physiol 2021; 320:C1000-C1012. [PMID: 33788629 DOI: 10.1152/ajpcell.00595.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity, especially visceral fat accumulation, increases the risk of type 2 diabetes (T2D). The purpose of this study was to investigate the impact of T2D on the pancreatic fat depot. Pancreatic fat pads from 17 partial pancreatectomized patients (PPP) were collected, pancreatic preadipocytes isolated, and in vitro differentiated. Patients were grouped using HbA1c into normal glucose tolerant (NGT), prediabetic (PD), and T2D. Transcriptome profiles of preadipocytes and adipocytes were assessed by RNAseq. Insulin sensitivity was estimated by quantifying AKT phosphorylation on Western blots. Lipogenic capacity was assessed with oil red O staining, lipolytic activity via fatty acid release. Secreted factors were measured using ELISA. Comparative transcriptome analysis of preadipocytes and adipocytes indicates defective upregulation of genes governing adipogenesis (NR1H3), lipogenesis (FASN, SCD, ELOVL6, and FADS1), and lipolysis (LIPE) during differentiation of cells from T2D-PPP. In addition, the ratio of leptin/adiponectin mRNA was higher in T2D than in NGT-PPP. Preadipocytes and adipocytes of NGT-PPP were more insulin sensitive than T2D-PPP cells in regard to AKT phosphorylation. Triglyceride accumulation was similar in NGT and T2D adipocytes. Despite a high expression of the receptors NPR1 and NPR2 in NGT and T2D adipocytes, lipolysis was stimulated by ANP 1.74-fold in NGT cells only. This stimulation was further increased by the PDE5 inhibitor dipyridamole (3.09-fold). Dipyridamole and forskolin increased lipolysis receptor independently 1.88-fold and 1.48-fold, respectively, solely in NGT cells. In conclusion, the metabolic status persistently affects differentiation and lipolysis of pancreatic adipocytes. These alterations could aggravate the development of T2D.
Collapse
Affiliation(s)
- Morgana Barroso Oquendo
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Dorothea Siegel-Axel
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Felicia Gerst
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Estela Lorza-Gil
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Anja Moller
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Robert Wagner
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Martin Heni
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany.,Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Susanne Ullrich
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| |
Collapse
|
17
|
Abstract
Adipose tissue depots in distinct anatomical locations mediate key aspects of metabolism, including energy storage, nutrient release, and thermogenesis. Although adipocytes make up more than 90% of adipose tissue volume, they represent less than 50% of its cellular content. Here, I review recent advances in genetic lineage tracing and transcriptomics that reveal the identities of the heterogeneous cell populations constituting mouse and human adipose tissues. In addition to mature adipocytes and their progenitors, these include endothelial and various immune cell types that together orchestrate adipose tissue development and functions. One salient finding is the identification of progenitor subtypes that can modulate adipogenic capacity through paracrine mechanisms. Another is the description of fate trajectories of monocyte/macrophages, which can respond maladaptively to nutritional and thermogenic stimuli, leading to metabolic disease. These studies have generated an extraordinary source of publicly available data that can be leveraged to explore commonalities and differences among experimental models, providing new insights into adipose tissues and their role in metabolic disease.
Collapse
Affiliation(s)
- Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA;
| |
Collapse
|
18
|
Al-Jaber H, Al-Mansoori L, Elrayess MA. GATA-3 as a Potential Therapeutic Target for Insulin Resistance and Type 2 Diabetes Mellitus. Curr Diabetes Rev 2021; 17:169-179. [PMID: 32628587 DOI: 10.2174/1573399816666200705210417] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 11/22/2022]
Abstract
Impaired adipogenesis plays an important role in the development of obesity-associated insulin resistance and type 2 diabetes as it leads to ectopic fat deposition. The anti-adipogenic transcription factor GATA-3 was identified as one of the potential molecular targets responsible for the impairment of adipogenesis. The expression of GATA-3 is higher in insulinresistant obese individuals compared to BMI-matched insulin-sensitive counterparts. Adipose tissue inflammation is a crucial mediator of this process. Hyperglycemia mediates the activation of the immune system, partially through upregulation of GATA- 3, causing exacerbation of the inflammatory state associated with obesity. This review discusses the evidence supporting the inhibition of GATA-3 as a useful therapeutic strategy in obesity-associated insulin resistance and type 2 diabetes, through up-regulation adipogenesis and amelioration of the immune response.
Collapse
Affiliation(s)
- Hend Al-Jaber
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | | |
Collapse
|
19
|
DeBari MK, Abbott RD. Adipose Tissue Fibrosis: Mechanisms, Models, and Importance. Int J Mol Sci 2020; 21:ijms21176030. [PMID: 32825788 PMCID: PMC7503256 DOI: 10.3390/ijms21176030] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
Increases in adipocyte volume and tissue mass due to obesity can result in inflammation, further dysregulation in adipose tissue function, and eventually adipose tissue fibrosis. Like other fibrotic diseases, adipose tissue fibrosis is the accumulation and increased production of extracellular matrix (ECM) proteins. Adipose tissue fibrosis has been linked to decreased insulin sensitivity, poor bariatric surgery outcomes, and difficulty in weight loss. With the rising rates of obesity, it is important to create accurate models for adipose tissue fibrosis to gain mechanistic insights and develop targeted treatments. This article discusses recent research in modeling adipose tissue fibrosis using in vivo and in vitro (2D and 3D) methods with considerations for biomaterial selections. Additionally, this article outlines the importance of adipose tissue in treating other fibrotic diseases and methods used to detect and characterize adipose tissue fibrosis.
Collapse
Affiliation(s)
- Megan K. DeBari
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Rosalyn D. Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Correspondence:
| |
Collapse
|
20
|
Fan R, You M, Toney AM, Kim J, Giraud D, Xian Y, Ye F, Gu L, Ramer-Tait AE, Chung S. Red Raspberry Polyphenols Attenuate High-Fat Diet-Driven Activation of NLRP3 Inflammasome and its Paracrine Suppression of Adipogenesis via Histone Modifications. Mol Nutr Food Res 2019; 64:e1900995. [PMID: 31786828 DOI: 10.1002/mnfr.201900995] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/18/2019] [Indexed: 12/28/2022]
Abstract
SCOPE The authors aim to investigate the mechanisms by which red raspberry (RR) polyphenolic fractions regulate obesity and inflammation with an emphasis on the crosstalk between adipose tissue macrophages (ATM) and adipocyte progenitors. METHODS AND RESULTS C57BL/6 male mice are fed either a high-fat (HF) diet or an HF diet supplemented with a RR polyphenolic fraction from whole fruit, pulp, or seed. Supplementation with pulp significantly increases energy expenditure and reduces HF-diet-induced obesity and insulin resistance. The pulp, and to a lesser extent, whole polyphenols, decreases the recruitment of ATM, activation of the nod-like receptor protein 3 (NLRP3) inflammasome, and adipocyte hypertrophy, which is associated with epigenetic modulation of adipogenesis (e.g., H3K27Ac, H3K9Ac). Results from an IL-1β reporter assay in J774 macrophages recapitulate the inhibitory role of RR polyphenols on NLRP3 inflammasome activation. Using conditioned media from macrophages, it is demonstrated that RR polyphenols reverse the IL-1β-mediated epigenetic suppression of H3K27Ac in adipocyte progenitor cells. CONCLUSIONS RR polyphenols from pulp and whole fruit serve as an inhibitor for NLRP3 inflammasome activation and an epigenetic modifier to regulate adipogenesis, which confers resistance against diet-induced obesity and metabolic dysfunction.
Collapse
Affiliation(s)
- Rong Fan
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE, 68583, USA
| | - Mikyoung You
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE, 68583, USA
| | - Ashley M Toney
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE, 68583, USA
| | - Judy Kim
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE, 68583, USA
| | - David Giraud
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE, 68583, USA
| | - Yibo Xian
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, 68583, USA
| | - Feng Ye
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA
| | - Liwei Gu
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA
| | - Amanda E Ramer-Tait
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, 68583, USA
| | - Soonkyu Chung
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE, 68583, USA
| |
Collapse
|
21
|
The Effect of Early Rounds of ex vivo Expansion and Cryopreservation on the Adipogenic Differentiation Capacity of Adipose-Derived Stromal/Stem Cells. Sci Rep 2019; 9:15943. [PMID: 31685852 PMCID: PMC6828715 DOI: 10.1038/s41598-019-52086-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/11/2019] [Indexed: 12/13/2022] Open
Abstract
Multipotent adipose-derived stromal/stem cells (ASCs) are candidates for use in cellular therapies for the treatment of a variety of conditions/diseases. Ex vivo expansion of freshly isolated ASCs may be necessary prior to clinical application to ensure that clinically relevant cell numbers are administered during treatment. In addition, cryopreserving cells at early passages allows for storage of freshly isolated cells for extended periods of time before expanding these cells for clinical usage. There are however several concerns that these laboratory-based procedures may alter the characteristics of the cells and in so doing decrease their regenerative potential. In this study we report on the impact of early rounds of cryopreservation (P0) and ex vivo expansion (P0 to P5) on the phenotypic characteristics and adipogenic differentiation potential of ASCs. Our results show that ASCs that upregulate CD36 expression during adipogenic differentiation gradually decrease with increasing expansion rounds. The consequent decrease in adipogenic differentiation capacity was evident in both gene expression and flow cytometry-based phenotypic studies. Successive rounds of expansion did not however alter cell surface marker expression of the cells. We also show that early cryopreservation of ASCs (at P0) does not affect the adipogenic differentiation potential of the cells.
Collapse
|
22
|
Wang Y, Lee MYK, Mak JCW, Ip MSM. Low-Frequency Intermittent Hypoxia Suppresses Subcutaneous Adipogenesis and Induces Macrophage Polarization in Lean Mice. Diabetes Metab J 2019; 43:659-674. [PMID: 31237128 PMCID: PMC6834831 DOI: 10.4093/dmj.2018.0196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/06/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The relationship between obstructive sleep apnoea (OSA) and metabolic disorders is complex and highly associated. The impairment of adipogenic capacity in pre-adipocytes may promote adipocyte hypertrophy and increase the risk of further metabolic dysfunction. We hypothesize that intermittent hypoxia (IH), as a pathophysiologic feature of OSA, may regulate adipogenesis by promoting macrophage polarization. METHODS Male C57BL/6N mice were exposed to either IH (240 seconds of 10% O₂ followed by 120 seconds of 21% O₂, i.e., 10 cycles/hour) or intermittent normoxia (IN) for 6 weeks. Stromal-vascular fractions derived from subcutaneous (SUB-SVF) and visceral (VIS-SVF) adipose tissues were cultured and differentiated. Conditioned media from cultured RAW 264.7 macrophages after air (Raw) or IH exposure (Raw-IH) were incubated with SUB-SVF during adipogenic differentiation. RESULTS Adipogenic differentiation of SUB-SVF but not VIS-SVF from IH-exposed mice was significantly downregulated in comparison with that derived from IN-exposed mice. IH-exposed mice compared to IN-exposed mice showed induction of hypertrophic adipocytes and increased preferential infiltration of M1 macrophages in subcutaneous adipose tissue (SAT) compared to visceral adipose tissue. Complementary in vitro analysis demonstrated that Raw-IH media significantly enhanced inhibition of adipogenesis of SUB-SVF compared to Raw media, in agreement with corresponding gene expression levels of differentiation-associated markers and adipogenic transcription factors. CONCLUSION Low frequency IH exposure impaired adipogenesis of SAT in lean mice, and macrophage polarization may be a potential mechanism for the impaired adipogenesis.
Collapse
Affiliation(s)
- Yan Wang
- Department of Medicine, The University of Hong Kong Li Ka Shing Faculty of Medicine, Hong Kong
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Mary Yuk Kwan Lee
- Department of Medicine, The University of Hong Kong Li Ka Shing Faculty of Medicine, Hong Kong
- Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong Li Ka Shing Faculty of Medicine, Hong Kong
| | - Judith Choi Wo Mak
- Department of Medicine, The University of Hong Kong Li Ka Shing Faculty of Medicine, Hong Kong
- Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong Li Ka Shing Faculty of Medicine, Hong Kong
- Department of Pharmacology & Pharmacy, The University of Hong Kong Li Ka Shing Faculty of Medicine, Hong Kong
| | - Mary Sau Man Ip
- Department of Medicine, The University of Hong Kong Li Ka Shing Faculty of Medicine, Hong Kong
- Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong Li Ka Shing Faculty of Medicine, Hong Kong.
| |
Collapse
|
23
|
Gerst F, Wagner R, Oquendo MB, Siegel-Axel D, Fritsche A, Heni M, Staiger H, Häring HU, Ullrich S. What role do fat cells play in pancreatic tissue? Mol Metab 2019; 25:1-10. [PMID: 31113756 PMCID: PMC6600604 DOI: 10.1016/j.molmet.2019.05.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/10/2019] [Accepted: 05/01/2019] [Indexed: 02/07/2023] Open
Abstract
Background It is now generally accepted that obesity is a major risk factor for type 2 diabetes mellitus (T2DM). Hepatic steatosis in particular, as well as visceral and ectopic fat accumulation within tissues, is associated with the development of the disease. We recently presented the first study on isolated human pancreatic adipocytes and their interaction with islets [Gerst, F., Wagner, R., Kaiser, G., Panse, M., Heni, M., Machann, J., et al., 2017. Metabolic crosstalk between fatty pancreas and fatty liver: effects on local inflammation and insulin secretion. Diabetologia 60(11):2240–2251.]. The results indicate that the function of adipocytes depends on the overall metabolic status in humans which, in turn, differentially affects islet hormone release. Scope of Review This review summarizes former and recent studies on factors derived from adipocytes and their effects on insulin-secreting β-cells, with particular emphasis on the human pancreas. The adipocyte secretome is discussed with a special focus on its influence on insulin secretion, β-cell survival and apoptotic β-cell death. Major Conclusions Human pancreatic adipocytes store lipids and release adipokines, metabolites, and pro-inflammatory molecules in response to the overall metabolic, humoral, and neuronal status. The differentially regulated adipocyte secretome impacts on endocrine function, i.e., insulin secretion, β-cell survival and death which interferes with glycemic control. This review attempts to explain why the extent of pancreatic steatosis is associated with reduced insulin secretion in some studies but not in others.
Collapse
Affiliation(s)
- Felicia Gerst
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Robert Wagner
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Morgana Barroso Oquendo
- German Center for Diabetes Research (DZD), Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Dorothea Siegel-Axel
- German Center for Diabetes Research (DZD), Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- German Center for Diabetes Research (DZD), Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Martin Heni
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Harald Staiger
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany; Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Susanne Ullrich
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany.
| |
Collapse
|
24
|
Lee MJ, Pickering RT, Shibad V, Wu Y, Karastergiou K, Jager M, Layne MD, Fried SK. Impaired Glucocorticoid Suppression of TGFβ Signaling in Human Omental Adipose Tissues Limits Adipogenesis and May Promote Fibrosis. Diabetes 2019; 68:587-597. [PMID: 30530781 PMCID: PMC6385749 DOI: 10.2337/db18-0955] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/26/2018] [Indexed: 12/18/2022]
Abstract
Visceral obesity is associated with insulin resistance and higher risk of type 2 diabetes and metabolic diseases. A limited ability of adipose tissues to remodel through the recruitment and differentiation of adipose stem cells (ASCs) is associated with adipose tissue inflammation and fibrosis and the metabolic syndrome. We show that the lower adipogenesis of omental (Om) compared with abdominal subcutaneous (Abdsc) ASCs was associated with greater secretion of TGFβ ligands that acted in an autocrine/paracrine loop to activate SMAD2 and suppress adipogenesis. Inhibition of TGFβ signaling rescued Om ASC differentiation. In Abdsc ASCs, low concentrations of dexamethasone suppressed TGFβ signaling and enhanced adipogenesis, at least in part by increasing TGFBR3 protein that can sequester TGFβ ligands. Om ASCs were resistant to these dexamethasone effects; recombinant TGFBR3 increased their differentiation. Pericellular fibrosis, a hallmark of dysfunctional adipose tissue, was greater in Om and correlated with higher level of tissue TGFβ signaling activity and lower ASC differentiation. We conclude that glucocorticoids restrain cell-autonomous TGFβ signaling in ASCs to facilitate adipogenesis and healthy remodeling in Abdsc and these processes are impaired in Om. Therapies directed at overcoming glucocorticoid resistance in visceral adipose tissue may improve remodeling and help prevent metabolic complications of visceral obesity.
Collapse
Affiliation(s)
- Mi-Jeong Lee
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| | - R Taylor Pickering
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| | - Varuna Shibad
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| | - Yuanyuan Wu
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| | - Kalypso Karastergiou
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| | - Mike Jager
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Matthew D Layne
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Susan K Fried
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| |
Collapse
|
25
|
Liu Y, Kongsuphol P, Chiam SY, Zhang QX, Gourikutty SBN, Saha S, Biswas SK, Ramadan Q. Adipose-on-a-chip: a dynamic microphysiological in vitro model of the human adipose for immune-metabolic analysis in type II diabetes. LAB ON A CHIP 2019; 19:241-253. [PMID: 30566152 DOI: 10.1039/c8lc00481a] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Infiltration of immune cells into adipose tissue is associated with chronic low-grade inflammation in obese individuals. To better understand the crosstalk between immune cells and adipocytes, in vivo-like in vitro models are required. Conventionally transwell culture plates are used for studying the adipocyte-immune cell interaction; however, the static culture nature of this approach falls short of closely recapitulating the physiological environment. Here we present a compartmentalized microfluidic co-culture system which provides a constant-rate of nutrient supply as well as waste removal, resembling the microvascular networks of the in vivo environment. Human adipocytes and U937 cells were co-cultured in close proximity in an enclosed system. The porous barrier between the adjacent compartments comprises an array of microchannels, which enables paracrine interaction between cells in adjacent compartments and improved perfusion-based long term cell feeding. Human pre-adipocytes were fully differentiated into adipocytes on the chip and remained viable for several weeks. Upon co-culturing with immune cells, adipocytes showed a tendency to develop insulin resistance. The immune-metabolic correlation has been studied by monitoring adiponectin and IL-6 expression, as well as glucose uptake upon treatment with insulin. Our microfluidic system can be potentially used to develop physiologically relevant adipose tissue models to study obesity-associated diseases such as insulin resistance and type 2 diabetes and therefore, facilitate drug development to treat these diseases.
Collapse
Affiliation(s)
- Yunxiao Liu
- Institute of Microelectronic, A* STAR (Agency for Science, Technology and Research), 2, Fusionopolis Way, #08-02, Innovis Tower, 138635 Singapore.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Mancuso P, Bouchard B. The Impact of Aging on Adipose Function and Adipokine Synthesis. Front Endocrinol (Lausanne) 2019; 10:137. [PMID: 30915034 PMCID: PMC6421296 DOI: 10.3389/fendo.2019.00137] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 02/13/2019] [Indexed: 02/04/2023] Open
Abstract
During the last 40 years, there has been a world-wide increase in both the prevalence of obesity and an increase in the number of persons over the age of 60 due to a decline in deaths from infectious disease and the nutrition transition in low and middle income nations. While the increase in the elderly population indicates improvements in global public health, this population may experience a diminished quality of life due to the negative impacts of obesity on age-associated inflammation. Aging alters adipose tissue composition and function resulting in insulin resistance and ectopic lipid storage. A reduction in brown adipose tissue activity, declining sex hormones levels, and abdominal adipose tissue expansion occur with advancing years through the redistribution of lipids from the subcutaneous to the visceral fat compartment. These changes in adipose tissue function and distribution influence the secretion of adipose tissue derived hormones, or adipokines, that promote a chronic state of low-grade systemic inflammation. Ultimately, obesity accelerates aging by enhancing inflammation and increasing the risk of age-associated diseases. The focus of this review is the impact of aging on adipose tissue distribution and function and how these effects influence the elaboration of pro and anti-inflammatory adipokines.
Collapse
Affiliation(s)
- Peter Mancuso
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
- Graduate Program in Immunology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Peter Mancuso
| | - Benjamin Bouchard
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
27
|
Bederman I, DiScenna A, Henderson L, Perez A, Klavanian J, Kovtun D, Collins O, Dunn J, Erokwu B, Flask CA, Drumm ML. Small adipose stores in cystic fibrosis mice are characterized by reduced cell volume, not cell number. Am J Physiol Gastrointest Liver Physiol 2018; 315:G943-G953. [PMID: 30188751 PMCID: PMC6336944 DOI: 10.1152/ajpgi.00096.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cystic fibrosis (CF) is a lethal genetic disorder that affects many organ systems of the body, including various endocrine and exocrine tissues. Health and survival positively associate with body mass, and as a consequence, CF clinical care includes high-fat, high-calorie diets to maintain and increase adipose tissue stores. Such strategies have been implemented without a clear understanding of the cause and effect relationship between body mass and patients' health. Here, we used CF mouse models, which display small adipose stores, to begin examining body fat as a prelude into mechanistic studies of low body growth in CF, so that optimal therapeutic strategies could be developed. We reasoned that low adiposity must result from reduced number and/or volume of adipocytes. To determine relative contribution of either mechanism, we quantified volume of intraperitoneal and subcutaneous adipocytes. We found smaller, but not fewer, adipocytes in CF compared with wild-type (WT) animals. Specifically, intraperitoneal CF adipocytes were one-half the volume of WT cells, whereas subcutaneous cells were less affected by the Cftr genotype. No differences were found in cell types between CF and WT adipose tissues. Adipose tissue CFTR mRNA was detected, and we found greater CFTR expression in intraperitoneal depots as compared with subcutaneous samples. RNA sequencing revealed that CF adipose tissue exhibited lower expression of several key genes of adipocyte function ( Lep, Pck1, Fas, Jun), consistent with low triglyceride storage. The data indicate that CF adipocytes contain fewer triglycerides than WT cells, and a role for CFTR in these cells is proposed. NEW & NOTEWORTHY Adipocytes in cystic fibrosis mice exhibit smaller size due to low triglyceride storage. Adipocyte cell number per fat pad is similar, implying triglyceride storage problem. The absence of CFTR function in adipose tissue has been proposed as a direct link to low triglyceride storage in cystic fibrosis.
Collapse
Affiliation(s)
- Ilya Bederman
- 1Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Alex DiScenna
- 1Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Leigh Henderson
- 2Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Aura Perez
- 1Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Jeannie Klavanian
- 2Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Daniel Kovtun
- 1Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Olivia Collins
- 1Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - John Dunn
- 1Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Bernadette Erokwu
- 3Department of Radiology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Christopher A. Flask
- 3Department of Radiology, School of Medicine, Case Western Reserve University, Cleveland, Ohio,4Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Mitchell L. Drumm
- 1Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio,2Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
28
|
Cytometric analysis of adipose tissue reveals increments of adipocyte progenitor cells after weight loss induced by bariatric surgery. Sci Rep 2018; 8:15203. [PMID: 30315279 PMCID: PMC6185966 DOI: 10.1038/s41598-018-33488-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/27/2018] [Indexed: 12/14/2022] Open
Abstract
Obesity-related comorbidities are, in large part, originated from the dysfunction of adipose tissue. Most of them revert after the normalization of body mass. Adipose tissue is essentially occupied by adipocytes. However, different populations of immunological cells and adipocyte precursor cells (AdPCs) are the main cellular components of tissue. During obesity, body fat depots acquire a low-level chronic inflammation and adipocytes increase in number and volume. Conversely, weight loss improves the inflammatory phenotype of adipose tissue immune cells and reduces the volume of adipocytes. Nevertheless, very little is known about the evolution of the human AdPCs reservoir. We have developed a flow cytometry-based methodology to simultaneously quantify the main cell populations of adipose tissue. Starting from this technical approach, we have studied human adipose tissue samples (visceral and subcutaneous) obtained at two different physiological situations: at morbid obesity and after bariatric surgery-induced weight loss. We report a considerable increase of the AdPCs reservoir after losing weight and several changes in the immune cells populations of adipose tissue (mast cells increase, neutrophils decrease and macrophages switch phenotype). No changes were observed for T-lymphocytes, which are discussed in the context of recent findings.
Collapse
|
29
|
Agrawal M, Yeo CR, Shabbir A, Chhay V, Silver DL, Magkos F, Vidal-Puig A, Toh SA. Fat storage-inducing transmembrane protein 2 (FIT2) is less abundant in type 2 diabetes, and regulates triglyceride accumulation and insulin sensitivity in adipocytes. FASEB J 2018; 33:430-440. [PMID: 30020828 DOI: 10.1096/fj.201701321rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Fat storage-inducing transmembrane protein 2 (FIT2) aids in partitioning of cellular triacylglycerol into lipid droplets. A genome-wide association study reported FITM2-R3H domain containing like-HNF4A locus to be associated with type 2 diabetes (T2DM) in East Asian populations. Mice with adipose tissue (AT)-specific FIT2 knockout exhibited lipodystrophic features, with reduced AT mass, insulin resistance, and greater inflammation in AT when fed a high-fat diet. The role of FIT2 in regulating human adipocyte function is not known. Here, we found FIT2 protein abundance is lower in subcutaneous and omental AT obtained from patients with T2DM compared with nondiabetic control subjects. Partial loss of FIT2 protein in primary human adipocytes attenuated their lipid storage capacity and induced insulin resistance. After palmitate treatment, triacylglycerol accumulation, insulin-induced Akt (Ser-473) phosphorylation, and insulin-stimulated glucose uptake were significantly reduced in FIT2 knockdown adipocytes compared with control cells. Gene expression of proinflammatory cytokines IL-18 and IL-6 and phosphorylation of the endoplasmic reticulum stress marker inositol-requiring enzyme 1α were greater in FIT2 knockdown adipocytes than in control cells. Our results show for the first time that FIT2 is associated with T2DM in humans and plays an integral role in maintaining metabolically healthy AT function.-Agrawal, M., Yeo, C. R., Shabbir, A., Chhay, V., Silver, D. L., Magkos, F., Vidal-Puig, A., Toh, S.-A. Fat storage-inducing transmembrane protein 2 (FIT2) is less abundant in type 2 diabetes, and regulates triglyceride accumulation and insulin sensitivity in adipocytes.
Collapse
Affiliation(s)
- Madhur Agrawal
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Chia Rou Yeo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Asim Shabbir
- Department of Surgery, National University Hospital, Singapore
| | - Vanna Chhay
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - David L Silver
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore
| | - Faidon Magkos
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Singapore Institute of Clinical Sciences (SICS), Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Antonio Vidal-Puig
- Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Sue-Anne Toh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Medicine, National University Health System, Singapore
| |
Collapse
|
30
|
Bharath LP, Ip BC, Nikolajczyk BS. Adaptive Immunity and Metabolic Health: Harmony Becomes Dissonant in Obesity and Aging. Compr Physiol 2017; 7:1307-1337. [PMID: 28915326 DOI: 10.1002/cphy.c160042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Adipose tissue (AT) is the primary energy reservoir organ, and thereby plays a critical role in energy homeostasis and regulation of metabolism. AT expands in response to chronic overnutrition or aging and becomes a major source of inflammation that has marked influence on systemic metabolism. The chronic, sterile inflammation that occurs in the AT during the development of obesity or in aging contributes to onset of devastating diseases such as insulin resistance, diabetes, and cardiovascular pathologies. Numerous studies have shown that inflammation in the visceral AT of humans and animals is a critical trigger for the development of metabolic syndrome. This work underscores the well-supported conclusion that the inflammatory immune response and metabolic pathways in the AT are tightly interwoven by multiple layers of relatively conserved mechanisms. During the development of diet-induced obesity or age-associated adiposity, cells of the innate and the adaptive immune systems infiltrate and proliferate in the AT. Macrophages, which dominate AT-associated immune cells in mouse models of obesity, but are less dominant in obese people, have been studied extensively. However, cells of the adaptive immune system, including T cells and B cells, contribute significantly to AT inflammation, perhaps more in humans than in mice. Lymphocytes regulate recruitment of innate immune cells into AT, and produce cytokines that influence the helpful-to-harmful inflammatory balance that, in turn, regulates organismal metabolism. This review describes inflammation, or more precisely, metabolic inflammation (metaflammation) with an eye toward the AT and the roles lymphocytes play in regulation of systemic metabolism during obesity and aging. © 2017 American Physiological Society. Compr Physiol 7:1307-1337, 2017.
Collapse
Affiliation(s)
- Leena P Bharath
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Blanche C Ip
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA.,Department of Molecular Pharmacology, Physiology and Biotechnology, Center of Biomedical Engineering, Brown University, Providence, Rhode Island, USA
| | | |
Collapse
|