1
|
Queiroz M, Sena CM. Perivascular adipose tissue: a central player in the triad of diabetes, obesity, and cardiovascular health. Cardiovasc Diabetol 2024; 23:455. [PMID: 39732729 DOI: 10.1186/s12933-024-02549-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024] Open
Abstract
Perivascular adipose tissue (PVAT) is a dynamic tissue that affects vascular function and cardiovascular health. The connection between PVAT, the immune system, obesity, and vascular disease is complex and plays a pivotal role in the pathogenesis of vascular diseases such as atherosclerosis, hypertension, and vascular inflammation. In cardiometabolic diseases, PVAT becomes a significant source of proflammatory adipokines, leading to increased infiltration of immune cells, in cardiometabolic diseases, PVAT becomes a significant source of proinflammatory adipokines, leading to increased infiltration of immune cells, promoting vascular smooth muscle cell proliferation and migrationpromoting vascular smooth muscle cell proliferation and migration. This exacerbates vascular dysfunction by impairing endothelial cell function and promoting endothelial activation. Dysregulated PVAT also contributes to hemodynamic alterations and hypertension through enhanced sympathetic nervous system activity and impaired vasodilatory capacity of PVAT-derived factors. Therapeutic interventions targeting key components of this interaction, such as modulating PVAT inflammation, restoring adipokine balance, and attenuating immune cell activation, hold promise for mitigating obesity-related vascular complications. Lifestyle interventions, pharmacological agents targeting inflammatory pathways, and surgical approaches aimed at reducing PVAT mass or improving adipose tissue function are potential therapeutic avenues for managing vascular diseases associated with obesity and PVAT dysfunction.
Collapse
Affiliation(s)
- Marcelo Queiroz
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Subunit 1, polo 3, Azinhaga de Santa Comba, Celas, 3000-548, Coimbra, Portugal
| | - Cristina M Sena
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Subunit 1, polo 3, Azinhaga de Santa Comba, Celas, 3000-548, Coimbra, Portugal.
| |
Collapse
|
2
|
De Siqueira MK, Li G, Zhao Y, Wang S, Ahn IS, Tamboline M, Hildreth AD, Larios J, Schcolnik-Cabrera A, Nouhi Z, Zhang Z, Tol MJ, Pandey V, Xu S, O'Sullivan TE, Mack JJ, Tontonoz P, Sallam T, Wohlschlegel JA, Hulea L, Xiao X, Yang X, Villanueva CJ. PPARγ-dependent remodeling of translational machinery in adipose progenitors is impaired in obesity. Cell Rep 2024; 43:114945. [PMID: 39579770 DOI: 10.1016/j.celrep.2024.114945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/14/2024] [Accepted: 10/17/2024] [Indexed: 11/25/2024] Open
Abstract
Adipose tissue regulates energy homeostasis and metabolic function, but its adaptability is impaired in obesity. In this study, we investigate the impact of acute PPARγ agonist treatment in obese mice and find significant transcriptional remodeling of cells in the stromal vascular fraction (SVF). Using single-cell RNA sequencing, we profile the SVF of inguinal and epididymal adipose tissue of obese mice following rosiglitazone treatment and find an induction of ribosomal factors in both progenitor and preadipocyte populations, while expression of ribosomal factors is reduced with obesity. Notably, the expression of a subset of ribosomal factors is directly regulated by PPARγ. Polysome profiling of the epididymal SVF shows that rosiglitazone promotes translational selectivity of mRNAs that encode pathways involved in adipogenesis and lipid metabolism. Inhibition of translation using a eukaryotic translation initiation factor 4A (eIF4A) inhibitor is sufficient in blocking adipogenesis. Our findings shed light on how PPARγ agonists promote adipose tissue plasticity in obesity.
Collapse
Affiliation(s)
- Mirian Krystel De Siqueira
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gaoyan Li
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yutian Zhao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Siqi Wang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - In Sook Ahn
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mikayla Tamboline
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90025, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90025, USA
| | - Andrew D Hildreth
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jakeline Larios
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alejandro Schcolnik-Cabrera
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, QC H1T 2M4, Canada; Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Zaynab Nouhi
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, QC H1T 2M4, Canada
| | - Zhengyi Zhang
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, Division of Cardiology, Los Angeles, Los Angeles, CA 90095, USA
| | - Marcus J Tol
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vijaya Pandey
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shili Xu
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90025, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90025, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90025, USA
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Julia J Mack
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, Division of Cardiology, Los Angeles, Los Angeles, CA 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tamer Sallam
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, Division of Cardiology, Los Angeles, Los Angeles, CA 90095, USA
| | - James A Wohlschlegel
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Laura Hulea
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, QC H1T 2M4, Canada; Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90025, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90025, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Claudio J Villanueva
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
3
|
Wang HY, Peng XM, Yang M, Weng Y, Yang X, Zhan D, Ning Q, Luo XP, Chen Y. C5aR1-positive adipocytes mediate non-shivering thermogenesis in neonatal mice. iScience 2024; 27:111261. [PMID: 39758991 PMCID: PMC11700647 DOI: 10.1016/j.isci.2024.111261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/25/2024] [Accepted: 10/23/2024] [Indexed: 01/07/2025] Open
Abstract
Brown adipose tissue (BAT) plays an important role in maintaining body temperature in newborn mammals; however, its mechanisms remain poorly understood. Here, we report the identification of a special population of brown adipose tissue-derived stromal cells (ASCs) in neonatal mice that highly express CD45 and can be differentiated into adipocytes with lower thermogenic ability. These CD45+ adipocytes also characteristically contained complement C5a receptor 1(C5aR1) on the cell membrane. C5ar1 deficiency in BAT resulted in an apparent immaturity of adipocytes and cold intolerance in neonatal mice. Mechanistically, loss of C5aR1 in these CD45+ brown adipocytes caused an increase in the secretion of plate factor four (PF4) from these cells, suppressing the maturity of neighboring brown adipocytes. Overall, our results indicated that the accumulation of C5aR1 positive brown adipocyte in neonatal BAT is essential for thermoregulation in newborn mice, which unveiled the regulatory mechanism of BAT-mediated thermogenesis in newborns.
Collapse
Affiliation(s)
- Huan-Yu Wang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Pediatric Genetic Metabolic and Endocrine Rare Diseases, Wuhan 430030, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue-Min Peng
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Min Yang
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Weng
- Department of Pediatrics, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Pediatric Genetic Metabolic and Endocrine Rare Diseases, Wuhan 430030, China
| | - Xi Yang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Pediatric Genetic Metabolic and Endocrine Rare Diseases, Wuhan 430030, China
| | - Di Zhan
- Department of Pediatrics, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Pediatric Genetic Metabolic and Endocrine Rare Diseases, Wuhan 430030, China
| | - Qin Ning
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Ping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Pediatric Genetic Metabolic and Endocrine Rare Diseases, Wuhan 430030, China
| | - Yong Chen
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
4
|
Li P, Ye H, Guo F, Zheng J, Shen W, Xie D, Shi S, Zhang Y, Fa Y, Zhao Z. Construction of cynomolgus monkey type 2 diabetes models by combining genetic prediction model with high-energy diet. Biochim Biophys Acta Mol Basis Dis 2024; 1871:167616. [PMID: 39672349 DOI: 10.1016/j.bbadis.2024.167616] [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: 08/21/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2D) is a significant health concern. Research using non-human primates, which develop T2D with similar symptoms and pancreatic changes as humans, is crucial but limited by long timelines and low success rates. RESULTS We targeted capture sequenced 61 normal and 81 T2D cynomolgus monkeys using a primer panel that captured 269 potential regulatory regions potentially associated with T2D in the cynomolgus monkey genome. 80 variants were identified to be associated with T2D and were used to construct a genetic prediction model. Among 8 machine learning algorithms tested, we found that the best prediction performance was achieve when the model using support vector machine with polynomial kernel as the machine learning algorithm (AUC = 0.933). Including age and sex in this model did not significantly improve the prediction performance. Using the genetic prediction model, we further screened 22 monkeys and found 13 were high risk while 9 were low risk. After feeding the 22 monkeys with high-energy food for 32 weeks, we found all the 9 low risk monkeys did not develop T2D while 4 out of 13 high risk monkeys (31 %) develop T2D. CONCLUSIONS This method greatly increased the success rate of establishing T2D monkey models while decreased the time needed compared to traditional methods. Therefore, we developed a new high-efficiency method to establish T2D monkey models by combining the genetic prediction model and high-energy diet, which will greatly contribute to the research on the clinical characteristics, pathogenesis, complications and potential new treatments.
Collapse
Affiliation(s)
- Ping Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Huahu Ye
- Academy of Military Medical Sciences, Beijing, China
| | - Feng Guo
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Jianhua Zheng
- Academy of Military Medical Sciences, Beijing, China
| | - Wenlong Shen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Dejian Xie
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Shu Shi
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Yan Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China.
| | - Yunzhi Fa
- Academy of Military Medical Sciences, Beijing, China
| | - Zhihu Zhao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
5
|
Huang Z, Yu X, Jiang Z, Tang G, Gao S, Xiang Y, Luo Y, Ye B, Li Y, Song P, Xin Y, Du M, Zhao J, Wang B. Neonatal vitamin A but not retinoic acid administration increases intramuscular adipocyte number in sheep by promoting vascularization. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 19:215-225. [PMID: 39635420 PMCID: PMC11615889 DOI: 10.1016/j.aninu.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/22/2024] [Accepted: 08/09/2024] [Indexed: 12/07/2024]
Abstract
This study investigated whether vitamin A (VA) administration during the neonatal stage could increase the number of intramuscular adipocytes in Hu sheep by promoting vascularity. A total of 56 newborn male Hu sheep were divided into four groups and received intramuscular injections of either 0, 7500 IU retinoic acid (RA), 7500 IU VA, or a combination of 7500 IU VA and 5 mg SU5416 (an angiogenic inhibitor), at 1, 7, 14, and 21 days of age. At 15 days of age, 6 sheep from each group were randomly selected and sacrificed for intramuscular adipogenic capacity analysis. The remaining 8 sheep in each group were raised until they were 8 months old. VA-treated sheep exhibited an increase in preadipocytes, elevated expression of adipogenic genes (CCAAT enhancer binding protein alpha [CEBPA] and CCAAT enhancer binding protein beta [CEBPB]) and angiogenic genes (vascular endothelial growth factor A [VEGFA]), and stromal vascular fraction cells in the longissimus dorsi (LD) muscle with enhanced adipogenic capacity (P < 0.05). These effects were entirely negated by SU5416. Upon slaughter, VA increased final weight, carcass weight, and average daily gain (P < 0.05) but did not affect feed intake at 21 to 32 weeks (P = 0.824). VA increased the number of intramuscular adipocytes in the LD and semitendinosus (ST) muscle (P < 0.05) without changing the adipocyte number of the omentum, perirenal and subcutaneous fats (P > 0.05). VA injections also increased intramuscular triglyceride (TG) content (P = 0.016) without changing the omentum fat weight or subcutaneous fat thickness (P > 0.05), but it did increase the perirenal fat weight (P = 0.011). Consistently, SU5416 mitigated the effects of VA on intramuscular TG content and adipocyte count, correlating with a decrease in vascularity. In contrast, RA injections didn't affect the intramuscular fat (P = 0.744) but reduced the TG content of the omentum and perirenal fat (P < 0.05). In conclusion, intramuscular injections of VA but not RA at the neonatal stage improved the growth performance of Hu sheep, increasing the number of intramuscular adipocytes and marbling by promoting angiogenesis.
Collapse
Affiliation(s)
- Zhongzuo Huang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiaoxiao Yu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zongyou Jiang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Gaojian Tang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shaoqi Gao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yifan Xiang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yicheng Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Boping Ye
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yating Li
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China
| | - Pengkang Song
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China
| | - Yu Xin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Min Du
- Laboratory of Nutrigenomics and Growth Biology, Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| | - Junxing Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China
| | - Bo Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
6
|
Kong J, Yao ZC, Stelzel JL, Yang YH, Chen J, Feng H, Schmidt C, Zhang C, Krishnan K, Chen L, Pan J, Ding K, Zhu Y, Li X, Doloff JC, Mao HQ, Reddy SK. Granular Nanofiber-Hydrogel Composite-Programmed Regenerative Inflammation and Adipose Tissue Formation. Adv Healthc Mater 2024:e2403094. [PMID: 39580666 DOI: 10.1002/adhm.202403094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/04/2024] [Indexed: 11/26/2024]
Abstract
The interplay between biomaterials and host immune responses critically determines outcomes in tissue restoration. Recent studies suggest that physicochemical properties of materials can dictate pro-regenerative versus pro-fibrotic responses and have begun to define the key immune cell types and signals governing these divergent effects. This emerging understanding enables the engineering of regenerative biomaterials capable of functional restoration in situ. An injectable nanofiber-hydrogel composite (NHC) microparticles are designed and constructed from cross-linked electrospun collagen nanofiber fragments surface-bonded to the hyaluronic acid hydrogel network via covalent conjugation during the cross-linking process. The collagen nanofiber fragments, acting as the structural reinforcement component, increased the overall storage modulus of the NHC to a level comparable to native soft tissues while maintaining a sufficiently high degree of porosity of the hydrogel phase to allow host cell infiltration following subcutaneous injection of the NHC microparticles. More importantly, the NHC promoted macrophage/monocyte infiltration, migration, and spreading, sustained cell recruitment over time, and enhanced the proangiogenic effect and recruitment of PDGFRα+ perivascular progenitor cells, leading to extensive adipose tissue remodeling. This study demonstrates the regenerative potential of the injectable NHC microgels as an off-the-shelf solution for devastating soft tissue losses.
Collapse
Affiliation(s)
- Jiayuan Kong
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Zhi-Cheng Yao
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jessica L Stelzel
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yueh-Hsun Yang
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Jeffrey Chen
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Hexiang Feng
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Collin Schmidt
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Chi Zhang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Kedar Krishnan
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Long Chen
- Department of Orthopaedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550000, P. R. China
| | - Jingwen Pan
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kailei Ding
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Yining Zhu
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Xiaowei Li
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joshua C Doloff
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Sashank K Reddy
- Translational Tissue Engineering Centre, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| |
Collapse
|
7
|
Chen ZJ, Das SS, Kar A, Lee SHT, Abuhanna KD, Alvarez M, Sukhatme MG, Gelev KZ, Heffel MG, Zhang Y, Avram O, Rahmani E, Sankararaman S, Heinonen S, Peltoniemi H, Halperin E, Pietiläinen KH, Luo C, Pajukanta P. Single-cell DNA methylome and 3D genome atlas of the human subcutaneous adipose tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.02.621694. [PMID: 39554055 PMCID: PMC11566006 DOI: 10.1101/2024.11.02.621694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Human subcutaneous adipose tissue (SAT) contains a diverse array of cell-types; however, the epigenomic landscape among the SAT cell-types has remained elusive. Our integrative analysis of single-cell resolution DNA methylation and chromatin conformation profiles (snm3C-seq), coupled with matching RNA expression (snRNA-seq), systematically cataloged the epigenomic, 3D topology, and transcriptomic dynamics across the SAT cell-types. We discovered that the SAT CG methylation (mCG) landscape is characterized by pronounced hyper-methylation in myeloid cells and hypo-methylation in adipocytes and adipose stem and progenitor cells (ASPCs), driving nearly half of the 705,063 detected differentially methylated regions (DMRs). In addition to the enriched cell-type-specific transcription factor binding motifs, we identified TET1 and DNMT3A as plausible candidates for regulating cell-type level mCG profiles. Furthermore, we observed that global mCG profiles closely correspond to SAT lineage, which is also reflected in cell-type-specific chromosome compartmentalization. Adipocytes, in particular, display significantly more short-range chromosomal interactions, facilitating the formation of complex local 3D genomic structures that regulate downstream transcriptomic activity, including those associated with adipogenesis. Finally, we discovered that variants in cell-type level DMRs and A compartments significantly predict and are enriched for variance explained in abdominal obesity. Together, our multimodal study characterizes human SAT epigenomic landscape at the cell-type resolution and links partitioned polygenic risk of abdominal obesity to SAT epigenome.
Collapse
|
8
|
Ellis CPS, Tero BW, Potts CM, Malka KT, Yang X, Hamilton J, Vary C, Khalil A, Liaw L. Cellular Characteristics and Protein Signatures of Human Adipose Tissues from Donors With or Without Advanced Coronary Artery Disease. Biomedicines 2024; 12:2453. [PMID: 39595019 PMCID: PMC11592159 DOI: 10.3390/biomedicines12112453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Perivascular adipose tissue (PVAT) exerts a paracrine effect on blood vessels and our objective was to understand PVAT molecular signatures related to cardiovascular disease. Methods: We studied two groups: those undergoing mitral valve repair/replacement (VR, n = 16) and coronary artery bypass graft (CABG, n = 38). VR donors did not have coronary artery disease, whereas CABG donors had advanced coronary artery disease. Clinical and tissue pathologies and proteomics from adipose tissue were assessed. Results: Donors undergoing VR had a lower body mass index (p = 0.01), HbA1C (p = 0.0023), and incidence of diabetes (p = 0.022) compared to CABG. VR donors were overall healthier, with higher cardiac function compared to CABG donors, based on ejection fraction. Although adipose histopathology between groups was not markedly different, PVAT had smaller and more adipocytes compared to subcutaneous adipose tissues. These differences were validated by whole specimen automated morphological analysis, and anisotropy analysis showed small (2.8-7.5 μm) and large (22.8-64.4 μm) scale differences between perivascular and subcutaneous adipose tissue from CABG donors, and small scale changes (2.8-7.5 μm) between perivascular and subcutaneous adipose tissue from VR donors. Distinct protein signatures in PVAT and subcutaneous adipose tissue include those involved in secretion, exosomes and vesicles, insulin resistance, and adipocyte identity. Comparing PVAT and subcutaneous adipose tissue from CABG donors, there were 82 significantly different proteins identified with log fold change ≥ 0.3 or ≤-0.3 (p < 0.05). Using this threshold, there were 36 differences when comparing PVAT and subcutaneous adipose tissue from VR donors, 58 differences when comparing PVAT from CABG or VR donors, and 55 when comparing subcutaneous adipose tissue from CABG vs. VR donors. Conclusions: Routine histopathology cannot differentiate between PVAT from donors with or without coronary artery disease, but multiscale anisotropy analysis discriminated between these populations. Our mass spectrometry analysis identified a cohort of proteins that distinguish between adipose depots, and are also associated with the presence or absence of coronary artery disease.
Collapse
Affiliation(s)
- Caitlin P. S. Ellis
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
| | - Benjamin W. Tero
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Christian M. Potts
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Kimberly T. Malka
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Xuehui Yang
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Joshua Hamilton
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
- CompuMAINE Lab, Department of Chemical and Biomedical Engineering, University of Maine, Orono, ME 04469, USA
| | - Calvin Vary
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
| | - Andre Khalil
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
- CompuMAINE Lab, Department of Chemical and Biomedical Engineering, University of Maine, Orono, ME 04469, USA
| | - Lucy Liaw
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
| |
Collapse
|
9
|
Yue S, Zhai G, Zhao S, Liang X, Liu Y, Zheng J, Chen X, Dong Y. The biphasic role of the infrapatellar fat pad in osteoarthritis. Biomed Pharmacother 2024; 179:117364. [PMID: 39226725 DOI: 10.1016/j.biopha.2024.117364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
Osteoarthritis (OA) is a progressive degenerative disease resulting in joint deterioration. It is a whole organ disease characterized by cartilage degeneration and varying degrees of synovitis, involving pathological changes in all joint tissues, such as cartilage, subchondral bone, ligaments, meniscus, synovium, and infrapatellar fat pad (IPFP). IPFP is the largest adipose tissue structure in the knee joint and is composed of fat cells, immune cells and blood vessels. Moreover, IPFP is located close to the cartilage and bone surface so that it may reduce the impact of loading and absorb forces generated through the knee joint, and may have a protective role in joint health. IPFP has been shown to release various cytokines and adipokines that play pro-inflammatory and pro-catabolic roles in cartilage, promoting OA progression. Intra-articular injections of IPFP-derived mesenchymal stem cells and exosomes have been shown to reduce pain and prevent OA progression in patients with knee OA. Previous studies have shown that IPFP has a biphasic effect on OA progression. This article reviews the latest research progress of IPFP, discusses the role and mechanism of IPFP in OA, provide new intervention strategies for the treatment of OA. This article will also discuss the handling of IPFP during the procedure of total knee arthroplasty.
Collapse
Affiliation(s)
- Songkai Yue
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Ganggang Zhai
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Siyu Zhao
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Xiaming Liang
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Yunke Liu
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Jia Zheng
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Xiaoyang Chen
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Yonghui Dong
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China.
| |
Collapse
|
10
|
Traynor S, Bhattacharya S, Batmanov K, Cheng L, Weller A, Moore N, Flesher C, Merrick D. Developmental regulation of dermal adipose tissue by BCL11b. Genes Dev 2024; 38:772-783. [PMID: 39266447 PMCID: PMC11444185 DOI: 10.1101/gad.351907.124] [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: 05/09/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024]
Abstract
The distinct anatomic environment in which adipose tissues arise during organogenesis is a principle determinant of their adult expansion capacity. Metabolic disease results from a deficiency in hyperplastic adipose expansion within the dermal/subcutaneous depot; thus, understanding the embryonic origins of dermal adipose is imperative. Using single-cell transcriptomics throughout murine embryogenesis, we characterized cell populations, including Bcl11b + cells, that regulate the development of dermal white adipose tissue (dWAT). We discovered that BCL11b expression modulates the Wnt signaling microenvironment to enable adipogenic differentiation in the dermal compartment. Subcutaneous and visceral adipose arises from a distinct population of Nefl + cells during embryonic organogenesis, whereas Pi16 + /Dpp4 + fibroadipogenic progenitors support obesity-stimulated hypertrophic expansion in the adult. Together, these results highlight the unique regulatory pathways used by anatomically distinct adipose depots, with important implications for human metabolic disease.
Collapse
Affiliation(s)
- Sarah Traynor
- Department of Medicine, Division of Endocrinology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Institute for Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Shashwati Bhattacharya
- Department of Medicine, Division of Endocrinology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Institute for Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Kirill Batmanov
- Department of Medicine, Division of Endocrinology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Institute for Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Lan Cheng
- Institute for Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Angela Weller
- Institute for Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Natalie Moore
- Department of Medicine, Division of Endocrinology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Institute for Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Carmen Flesher
- Department of Medicine, Division of Endocrinology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Institute for Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - David Merrick
- Department of Medicine, Division of Endocrinology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Institute for Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
11
|
Mao L, Lu J, Hou Y, Nie T. Directly targeting PRDM16 in thermogenic adipose tissue to treat obesity and its related metabolic diseases. Front Endocrinol (Lausanne) 2024; 15:1458848. [PMID: 39351529 PMCID: PMC11439700 DOI: 10.3389/fendo.2024.1458848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
Obesity is increasing globally and is closely associated with a range of metabolic disorders, including metabolic associated fatty liver disease, diabetes, and cardiovascular diseases. An effective strategy to combat obesity involves stimulating brown and beige adipocyte thermogenesis, which significantly enhances energy expenditure. Recent research has underscored the vital role of PRDM16 in the development and functionality of thermogenic adipocytes. Consequently, PRDM16 has been identified as a potential therapeutic target for obesity and its related metabolic disorders. This review comprehensively examines various studies that focus on combating obesity by directly targeting PRDM16 in adipose tissue.
Collapse
Affiliation(s)
- Liufeng Mao
- The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jinli Lu
- The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yunliang Hou
- The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tao Nie
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
12
|
Stephens CJ, Kobayashi R, Berry DC, Butcher JT. The Role of Matrix Stiffness And Viscosity on Lipid Phenotype And Fat Lineage Potential. Tissue Eng Part A 2024. [PMID: 39165245 DOI: 10.1089/ten.tea.2024.0149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024] Open
Abstract
Autologous fat transfer is a common procedure that patients undergo to rejuvenate large soft tissue defects. However, these surgeries are complicated by limited tissue sources, donor-site morbidity, and necrosis. While the biofabrication of fat tissue can serve as a clinical option for reconstructive surgery, the influence of matrix mechanics, specifically stiffness and viscosity, on adipogenesis requires further elucidation. Additionally, the effects of these mechanical parameters on metabolic and thermogenic fat potential have yet to be investigated. In this study, gelatin methacryloyl (GelMA) polymers with varying degrees of methacrylation (DoM) were fabricated to create matrices with different stiffnesses and viscosities. Human adipose-derived mesenchymal stem cells were then encapsulated in mechanically tunable GelMA and underwent adipogenesis to investigate the effects of matrix mechanics on lipid phenotype and fat potential. Mechanical testing confirmed that GelMA stiffness was regulated by DoM and weight composition, whereas viscosity was determined by the latter. Further work revealed that while lipid phenotype became more enriched as matrix stiffness and viscosity declined, the potential toward metabolic and thermogenic fat appeared to be more viscous dependent rather than stiffness dependent. In addition, fatty acid binding protein 4 and uncoupling protein 1 gene expression exhibited viscous-dependent behavior despite comparable levels of peroxisome proliferator-activated receptor gamma. However, despite the superior role of viscosity, lipid quantity and mitochondrial abundance demonstrated stiffness-dependent behavior. Overall, this work revealed that matrix viscosity played a more superior role than stiffness in driving adipogenesis and distinguishing between metabolic and thermogenic fat potential. Ultimately, this differentiation in fat production is important for engineering ideal adipose tissue for large soft tissue defects.
Collapse
Affiliation(s)
- Chelsea J Stephens
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| | - Reina Kobayashi
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| | - Daniel C Berry
- College of Human Ecology, Division of Nutrition Science, Cornell University, Ithaca, New York, USA
| | - Jonathan T Butcher
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| |
Collapse
|
13
|
Li Y, Chen Z, Xiao Y, Li X. Cross-talks between perivascular adipose tissue and neighbors: multifaceted nature of nereids. Front Pharmacol 2024; 15:1442086. [PMID: 39156105 PMCID: PMC11327032 DOI: 10.3389/fphar.2024.1442086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 07/24/2024] [Indexed: 08/20/2024] Open
Abstract
Perivascular adipose tissue (PVAT) is a unique fat depot surrounding blood vessels and plays a vital role in the progression of vascular remodeling and dysfunction. PVAT exhibits remarkable differences in structure, phenotype, origin, and secretome across anatomical locations. The proximity of PVAT to neighboring vascular beds favors a niche for bidirectional communication between adipocytes and vascular smooth muscle cells, endothelial cells, and immune cells. In this review, we update our understanding of PVAT's regional differences and provide a comprehensive exploration of how these differences impact cross-talks between PVAT and the vascular wall. Different PVAT depots show different degrees of vasoprotective function and resilience to pathological changes such as obesity and vasculopathies, shaping multifaceted interactions between PVAT depots and adjacent vasculatures. The depot-specific resilience may lead to innovative strategies to manage cardiometabolic disorders.
Collapse
Affiliation(s)
- Yujuan Li
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, China
- (R & D Center) Laboratory for Drug Discovery from Natural Resource, Macau University of Science and Technology, Macau, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Zhang Chen
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, China
- (R & D Center) Laboratory for Drug Discovery from Natural Resource, Macau University of Science and Technology, Macau, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Ying Xiao
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Xinzhi Li
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, China
- (R & D Center) Laboratory for Drug Discovery from Natural Resource, Macau University of Science and Technology, Macau, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| |
Collapse
|
14
|
Rendon CJ, Sempere L, Lauver A, Watts SW, Contreras GA. Anatomical location, sex, and age modulate adipocyte progenitor populations in perivascular adipose tissues. Front Physiol 2024; 15:1411218. [PMID: 39072214 PMCID: PMC11282503 DOI: 10.3389/fphys.2024.1411218] [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: 04/02/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024] Open
Abstract
Perivascular adipose tissue (PVAT) regulates vascular function due to its capacity to synthesize vasoactive products and its mechanical properties. PVATs most abundant cells are adipocytes, and their populations are maintained by the maturation of adipocyte progenitor cells (APC), which may play a pivotal role in the pathogenesis of cardiovascular diseases. However, the distribution of APC within PVAT depots, their potential variation in spatial location, and the influence of sex and age on their abundance remain unknown. We hypothesize that APC abundance in PVAT is affected by location, age, sex and that APC subtypes have specific spatial distributions. PVAT from thoracic and abdominal aorta, and mesenteric arteries, and AT from interscapular, gonadal, and subcutaneous depots from 13-week and 30-week-old females and males Pdgfrα-CreERT2 x LSL-tdTomato mice (n = 28) were analyzed. Abdominal aorta PVAT had fewer progenitors than mesenteric PVAT and gonadal AT. Aging reduced the abundance of APC in the thoracic aorta but increased their numbers in mesenteric PVAT. Females had more APC than males in mesenteric PVAT and gonadal AT depots. APC exhibited unique spatial distribution in the aorta and mesenteric PVAT where they localized neighboring vasa vasorum and arteries. APC subtypes (APC1, APC2, APC3, diff APC) were identified in all PVAT depots. Thoracic aorta PVAT APC3 were located in the adventitia while diff APC were in the parenchyma. This study identified variability in APC populations based on depot, age, and sex. The distinctive spatial distribution and the presence of diverse APC subtypes suggest that they may contribute differently to cardiovascular diseases-induced PVAT remodeling.
Collapse
Affiliation(s)
- C. Javier Rendon
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Lorenzo Sempere
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, United States
| | - Adam Lauver
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Stephanie W. Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - G. Andres Contreras
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
15
|
Lin T, Mohammad A, Kolonin MG, Eckel-Mahan KL. Mechanisms and metabolic consequences of adipocyte progenitor replicative senescence. IMMUNOMETABOLISM (COBHAM, SURREY) 2024; 6:e00046. [PMID: 39211801 PMCID: PMC11356692 DOI: 10.1097/in9.0000000000000046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
In recent decades, obesity has become a worldwide epidemic. As a result, the importance of adipose tissue (AT) as a metabolically active storage depot for lipids and a key mediator of body-wide metabolism and energy balance has been increasingly recognized. Emerging from the studies of AT in metabolic disease is a recognition of the importance of the adipocyte progenitor cell (APC) population of AT being the gatekeeper of adipocyte function. APCs have the capability to self-renew and undergo adipogenesis to propagate new adipocytes capable of lipid storage, which is important for maintaining a healthy fat pad, devoid of dysfunctional lipid droplet hypertrophy, inflammation, and fibrosis, which is linked to metabolic diseases, including type 2 diabetes. Like other dividing cells, APCs are at risk for undergoing cell senescence, a state of irreversible cell proliferation arrest that occurs under a variety of stress conditions, including DNA damage and telomere attrition. APC proliferation is controlled by a variety of factors, including paracrine and endocrine factors, quality and timing of energy intake, and the circadian clock system. Therefore, alteration in any of the underlying signaling pathways resulting in excessive proliferation of APCs can lead to premature APC senescence. Better understanding of APCs senescence mechanisms will lead to new interventions extending metabolic health.
Collapse
Affiliation(s)
- Tonghui Lin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Aftab Mohammad
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- Molecular and Translational Biology Program, MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kristin L. Eckel-Mahan
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- Molecular and Translational Biology Program, MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
16
|
Liu Z, Wang J, Tian P, Liu Y, Xing L, Fu C, Huang X, Liu P. Sodium-glucose cotransporter 1 promotes the biofunctions of perivascular preadipocytes mediated by Akt/mTOR/p70S6K signaling pathway. Am J Physiol Cell Physiol 2024; 326:C1611-C1624. [PMID: 38646789 PMCID: PMC11371362 DOI: 10.1152/ajpcell.00606.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/23/2024]
Abstract
The influence of SGLT-1 on perivascular preadipocytes (PVPACs) and vascular remodeling is not well understood. This study aimed to elucidate the role and mechanism of SGLT-1-mediated PVPACs bioactivity. PVPACs were cultured in vitro and applied ex vivo to the carotid arteries of mice using a lentivirus-based thermosensitive in situ gel (TISG). The groups were treated with Lv-SGLT1 (lentiviral vector, overexpression), Lv-siSGLT1 (RNA interference, knockdown), or specific signaling pathway inhibitors. Assays were conducted to assess changes in cell proliferation, apoptosis, glucose uptake, adipogenic differentiation, and vascular remodeling in the PVPACs. Protein expression was analyzed by Western blotting, immunocytochemistry, and/or immunohistochemistry. The methyl thiazolyl tetrazolium (MTT) assay and Hoechst 33342 staining indicated that SGLT-1 overexpression significantly promoted PVPACs proliferation and inhibited apoptosis in vitro. Conversely, SGLT-1 knockdown exerted the opposite effect. Oil Red O staining revealed that SGLT-1 overexpression facilitated adipogenic differentiation, while its inhibition mitigated these effects. 3H-labeled glucose uptake experiments demonstrated that SGLT-1 overexpression enhanced glucose uptake by PVPACs, whereas RNA interference-mediated SGLT-1 inhibition had no significant effect on glucose uptake. Moreover, RT-qPCR, Western blotting, and immunofluorescence analyses revealed that SGLT-1 overexpression upregulated FABP4 and VEGF-A levels and activated the Akt/mTOR/p70S6K signaling pathway, whereas SGLT-1 knockdown produced the opposite effects. In vivo studies corroborated these findings and indicated that SGLT-1 overexpression facilitated carotid artery remodeling. Our study demonstrates that SGLT-1 activation of the Akt/mTOR/p70S6K signaling pathway promotes PVPACs proliferation, adipogenesis, glucose uptake, glucolipid metabolism, and vascular remodeling.NEW & NOTEWORTHY SGLT-1 is expressed in PVPACs and can affect preadipocyte glucolipid metabolism and vascular remodeling. SGLT-1 promotes the biofunctions of PVPACs mediated by Akt/mTOR/p70S6K signaling pathway. Compared with caudal vein or intraperitoneal injection, the external application of lentivirus-based thermal gel around the carotid artery is an innovative attempt at vascular remodeling model, it may effectively avoid the transfection of lentiviral vector into the whole body of mice and the adverse effect on experimental results.
Collapse
Affiliation(s)
- Zhiquan Liu
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, China
- Division of Life Sciences and Medicine, Department of Cardiology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Jiayu Wang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, China
| | - Peiqing Tian
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, China
| | - Yixuan Liu
- Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Liyun Xing
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, China
| | - Caihua Fu
- Department of Cardiology, Jinan Central Hospital Affiliated Shandong University, Jinan, China
| | - Xianwei Huang
- Department of Emergency, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Ping Liu
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
17
|
Shi Z, Xiong S, Hu R, Wang Z, Park J, Qian Y, Wang J, Bhalla P, Velupally N, Song Q, Song Z, Jeon MS, Zhang KK, Xie L, Layden BT, Ong SG, Jiang Y. The Notch-PDGFRβ axis suppresses brown adipocyte progenitor differentiation in early post-natal mice. Dev Cell 2024; 59:1233-1251.e5. [PMID: 38569546 DOI: 10.1016/j.devcel.2024.03.012] [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: 05/23/2023] [Revised: 12/08/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
De novo brown adipogenesis holds potential in combating the epidemics of obesity and diabetes. However, the identity of brown adipocyte progenitor cells (APCs) and their regulation have not been extensively explored. Here, through in vivo lineage tracing and mouse modeling, we observed that platelet-derived growth factor receptor beta (PDGFRβ)+ pericytes give rise to developmental brown adipocytes but not to those in adult homeostasis. By contrast, T-box 18 (TBX18)+ pericytes contribute to brown adipogenesis throughout both developmental and adult stages, though in a depot-specific manner. Mechanistically, Notch inhibition in PDGFRβ+ pericytes promotes brown adipogenesis by downregulating PDGFRβ. Furthermore, inhibition of Notch signaling in PDGFRβ+ pericytes mitigates high-fat, high-sucrose (HFHS)-induced glucose and metabolic impairment in mice during their development and juvenile phases. Collectively, these findings show that the Notch/PDGFRβ axis negatively regulates developmental brown adipogenesis, and its repression promotes brown adipose tissue expansion and improves metabolic health.
Collapse
Affiliation(s)
- Zuoxiao Shi
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Shaolei Xiong
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ruoci Hu
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zilai Wang
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Jooman Park
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Yanyu Qian
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Jaden Wang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Pratibha Bhalla
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Nipun Velupally
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Qing Song
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Minsun Stacey Jeon
- Texas A&M Health Science Center, Institute of Biosciences and Technology, Houston, TX 77030, USA
| | - Ke Kurt Zhang
- Texas A&M Health Science Center, Institute of Biosciences and Technology, Houston, TX 77030, USA
| | - Linlin Xie
- Department of Nutrition, Texas A&M University, College Station, TX 77845, USA
| | - Brian T Layden
- Division of Endocrinology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Jesse Brown Medical VA Medical Center, Chicago, IL 60612, USA
| | - Sang-Ging Ong
- Department of Pharmacology and Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Yuwei Jiang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA; Division of Endocrinology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
18
|
Wang Y, Wang X, Chen Y, Zhang Y, Zhen X, Tao S, Dou J, Li P, Jiang G. Perivascular fat tissue and vascular aging: A sword and a shield. Pharmacol Res 2024; 203:107140. [PMID: 38513826 DOI: 10.1016/j.phrs.2024.107140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 02/16/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
The understanding of the function of perivascular adipose tissue (PVAT) in vascular aging has significantly changed due to the increasing amount of information regarding its biology. Adipose tissue surrounding blood vessels is increasingly recognized as a key regulator of vascular disorders. It has significant endocrine and paracrine effects on the vasculature and is mediated by the production of a variety of bioactive chemicals. It also participates in a number of pathological regulatory processes, including oxidative stress, immunological inflammation, lipid metabolism, vasoconstriction, and dilation. Mechanisms of homeostasis and interactions between cells at the local level tightly regulate the function and secretory repertoire of PVAT, which can become dysregulated during vascular aging. The PVAT secretion group changes from being reducing inflammation and lowering cholesterol to increasing inflammation and increasing cholesterol in response to systemic or local inflammation and insulin resistance. In addition, the interaction between the PVAT and the vasculature is reciprocal, and the biological processes of PVAT are directly influenced by the pertinent indicators of vascular aging. The architectural and biological traits of PVAT, the molecular mechanism of crosstalk between PVAT and vascular aging, and the clinical correlation of vascular age-related disorders are all summarized in this review. In addition, this paper aims to elucidate and evaluate the potential benefits of therapeutically targeting PVAT in the context of mitigating vascular aging. Furthermore, it will discuss the latest advancements in technology used for targeting PVAT.
Collapse
Affiliation(s)
- Yan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xianmin Wang
- Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Xinjiang 830000, China
| | - Yang Chen
- School of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang 830011, China
| | - Yuelin Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xianjie Zhen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Siyu Tao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jinfang Dou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Peng Li
- Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Xinjiang 830000, China
| | - Guangjian Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; School of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang 830011, China.
| |
Collapse
|
19
|
Thompson JM, Watts SW, Terrian L, Contreras GA, Rockwell C, Rendon CJ, Wabel E, Lockwood L, Bhattacharya S, Nault R. A cell atlas of thoracic aortic perivascular adipose tissue: a focus on mechanotransducers. Am J Physiol Heart Circ Physiol 2024; 326:H1252-H1265. [PMID: 38517229 PMCID: PMC11380965 DOI: 10.1152/ajpheart.00040.2024] [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: 01/23/2024] [Revised: 03/05/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
Perivascular adipose tissue (PVAT) is increasingly recognized for its function in mechanotransduction. However, major gaps remain in our understanding of the cells present in PVAT, as well as how different cells contribute to mechanotransduction. We hypothesized that snRNA-seq would reveal the expression of mechanotransducers, and test one (PIEZO1) to illustrate the expression and functional agreement between single-nuclei RNA sequencing (snRNA-seq) and physiological measurements. To contrast two brown tissues, subscapular brown adipose tissue (BAT) was also examined. We used snRNA-seq of the thoracic aorta PVAT (taPVAT) and BAT from male Dahl salt-sensitive (Dahl SS) rats to investigate cell-specific expression mechanotransducers. Localization and function of the mechanostransducer PIEZO1 were further examined using immunohistochemistry (IHC) and RNAscope, as well as pharmacological antagonism. Approximately 30,000 nuclei from taPVAT and BAT each were characterized by snRNA-seq, identifying eight major cell types expected and one unexpected (nuclei with oligodendrocyte marker genes). Cell-specific differential gene expression analysis between taPVAT and BAT identified up to 511 genes (adipocytes) with many (≥20%) being unique to individual cell types. Piezo1 was the most highly, widely expressed mechanotransducer. The presence of PIEZO1 in the PVAT but not the adventitia was confirmed by RNAscope and IHC in male and female rats. Importantly, antagonism of PIEZO1 by GsMTX4 impaired the PVAT's ability to hold tension. Collectively, the cell compositions of taPVAT and BAT are highly similar, and PIEZO1 is likely a mechanotransducer in taPVAT.NEW & NOTEWORTHY This study describes the atlas of cells in the thoracic aorta perivascular adipose tissue (taPVAT) of the Dahl-SS rat, an important hypertension model. We show that mechanotransducers are widely expressed in these cells. Moreover, PIEZO1 expression is shown to be restricted to the taPVAT and is functionally implicated in stress relaxation. These data will serve as the foundation for future studies investigating the role of taPVAT in this model of hypertensive disease.
Collapse
Affiliation(s)
- Janice M Thompson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Leah Terrian
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, United States
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, United States
| | - Cheryl Rockwell
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - C Javier Rendon
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, United States
| | - Emma Wabel
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Lizbeth Lockwood
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Sudin Bhattacharya
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Rance Nault
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, United States
| |
Collapse
|
20
|
Wang A, Dong S, Liu B, Liu D, Zou M, Han Y, Yang L, Wang Y. The role of RUNX1/NF-κB in regulating PVAT inflammation in aortic dissection. Sci Rep 2024; 14:9960. [PMID: 38693222 PMCID: PMC11063189 DOI: 10.1038/s41598-024-60737-9] [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: 11/11/2023] [Accepted: 04/26/2024] [Indexed: 05/03/2024] Open
Abstract
The pathogenesis of aortic dissection (AD), an aortic disease associated with high mortality, involves significant vascular inflammatory infiltration. However, the precise relationship between perivascular adipose tissue (PVAT) and aortic dissection remains incompletely understood. The objective of this study is to investigate the role of PVAT inflammation in the pathogenesis of aortic dissection and identify novel therapeutic targets for this disease. The mouse model of aortic dissection was established in this study through intraperitoneal injection of Ang II and administration of BAPN in drinking water. Additionally, control groups were established at different time points including the 2-week group, 3-week group, and 4-week group. qPCR and immunohistochemistry techniques were employed to detect the expression of inflammatory markers and RUNX1 in PVAT surrounding the thoracic aorta in mice. Additionally, an aortic dissection model was established using RUNX1 knockout mice, and the aforementioned indicators were assessed. The 3T3-L1 cells were induced to differentiate into mature adipocytes in vitro, followed by lentivirus transfection for the knockdown or overexpression of RUNX1. The study aimed to investigate the potential cell-to-cell interactions by co-culturing 3T3-L1 cells with A7r5 or RAW264.7 cells. Subsequently, human aortic PVAT samples were obtained through clinical surgery and the aforementioned indicators were detected. In comparison to the control group, the aortic dissection model group exhibited decreased expression of MMP-2 and NF-κB in PVAT, while TNF-α and RUNX1 expression increased. Suppression of RUNX1 expression resulted in increased MMP-2 and NF-κB expression in PVAT, along with decreased TNF-α expression. Overexpression of RUNX1 upregulated the expression levels of NF-Κb, MMP-2, and TNF-α in adipocytes, whereas knockdown of RUNX1 exerted an opposite effect. Macrophages co-cultured with adipocytes overexpressing RUNX1 exhibited enhanced CD86 expression, while vascular smooth muscle cells co-cultured with these adipocytes showed reduced α-SMA expression. In human samples, there was an increase in both RUNX1 and MMP-2 expression levels, accompanied by a decrease in TNF-α and NF-Κb expression. The presence of aortic dissection is accompanied by evident inflammatory alterations in the PVAT, and this phenomenon appears to be associated with the involvement of RUNX1. It is plausible that the regulation of PVAT's inflammatory changes by RUNX1/NF-κB signaling pathway plays a role in the pathogenesis of aortic dissection.
Collapse
Affiliation(s)
- Ao Wang
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Shengjun Dong
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Baohui Liu
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Dianxiao Liu
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Mingrui Zou
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Yuexin Han
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Lijuan Yang
- Department of Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China.
| | - Yujiu Wang
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China.
| |
Collapse
|
21
|
Peng Y, Zhao L, Li M, Liu Y, Shi Y, Zhang J. Plasticity of Adipose Tissues: Interconversion among White, Brown, and Beige Fat and Its Role in Energy Homeostasis. Biomolecules 2024; 14:483. [PMID: 38672499 PMCID: PMC11048349 DOI: 10.3390/biom14040483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Obesity, characterized by the excessive accumulation of adipose tissue, has emerged as a major public health concern worldwide. To develop effective strategies for treating obesity, it is essential to comprehend the biological properties of different adipose tissue types and their respective roles in maintaining energy balance. Adipose tissue serves as a crucial organ for energy storage and metabolism in the human body, with functions extending beyond simple fat storage to encompass the regulation of energy homeostasis and the secretion of endocrine factors. This review provides an overview of the key characteristics, functional differences, and interconversion processes among white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue. Moreover, it delves into the molecular mechanisms and recent research advancements concerning the browning of WAT, activation of BAT, and whitening of BAT. Although targeting adipose tissue metabolism holds promise as a potential approach for obesity treatment, further investigations are necessary to unravel the intricate biological features of various adipose tissue types and elucidate the molecular pathways governing their interconversion. Such research endeavors will pave the way for the development of more efficient and targeted therapeutic interventions in the fight against obesity.
Collapse
Affiliation(s)
| | | | | | | | | | - Jian Zhang
- School of Bioengineering, Zunyi Medical University, Zhuhai 519000, China; (Y.P.); (L.Z.); (M.L.); (Y.L.); (Y.S.)
| |
Collapse
|
22
|
Shah R, Zhong J, Massier L, Tanriverdi K, Hwang SJ, Haessler J, Nayor M, Zhao S, Perry AS, Wilkins JT, Shadyab AH, Manson JE, Martin L, Levy D, Kooperberg C, Freedman JE, Rydén M, Murthy VL. Targeted Proteomics Reveals Functional Targets for Early Diabetes Susceptibility in Young Adults. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004192. [PMID: 38323454 PMCID: PMC10940209 DOI: 10.1161/circgen.123.004192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 11/05/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND The circulating proteome may encode early pathways of diabetes susceptibility in young adults for surveillance and intervention. Here, we define proteomic correlates of tissue phenotypes and diabetes in young adults. METHODS We used penalized models and principal components analysis to generate parsimonious proteomic signatures of diabetes susceptibility based on phenotypes and on diabetes diagnosis across 184 proteins in >2000 young adults in the CARDIA (Coronary Artery Risk Development in Young Adults study; mean age, 32 years; 44% women; 43% Black; mean body mass index, 25.6±4.9 kg/m2), with validation against diabetes in >1800 individuals in the FHS (Framingham Heart Study) and WHI (Women's Health Initiative). RESULTS In 184 proteins in >2000 young adults in CARDIA, we identified 2 proteotypes of diabetes susceptibility-a proinflammatory fat proteotype (visceral fat, liver fat, inflammatory biomarkers) and a muscularity proteotype (muscle mass), linked to diabetes in CARDIA and WHI/FHS. These proteotypes specified broad mechanisms of early diabetes pathogenesis, including transorgan communication, hepatic and skeletal muscle stress responses, vascular inflammation and hemostasis, fibrosis, and renal injury. Using human adipose tissue single cell/nuclear RNA-seq, we demonstrate expression at transcriptional level for implicated proteins across adipocytes and nonadipocyte cell types (eg, fibroadipogenic precursors, immune and vascular cells). Using functional assays in human adipose tissue, we demonstrate the association of expression of genes encoding these implicated proteins with adipose tissue metabolism, inflammation, and insulin resistance. CONCLUSIONS A multifaceted discovery effort uniting proteomics, underlying clinical susceptibility phenotypes, and tissue expression patterns may uncover potentially novel functional biomarkers of early diabetes susceptibility in young adults for future mechanistic evaluation.
Collapse
Affiliation(s)
- Ravi Shah
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | - Jiawei Zhong
- Dept of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Lucas Massier
- Dept of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Kahraman Tanriverdi
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | - Shih-Jen Hwang
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Matthew Nayor
- Sections of Preventive Medicine & Epidemiology & Cardiovascular Medicine, Dept of Medicine, Dept of Epidemiology, Boston University Schools of Medicine & Public Health, Boston, MA & Framingham Heart Study, Framingham, MA
| | | | - Andrew S. Perry
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | | | - Aladdin H. Shadyab
- Herbert Wertheim School of Public Health & Human Longevity Science, Univ of California, San Diego, La Jolla, CA
| | - JoAnn E. Manson
- Dept of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Lisa Martin
- George Washington Univ School of Medicine & Health Sciences
| | - Daniel Levy
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Jane E. Freedman
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | - Mikael Rydén
- Dept of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
23
|
Jeon YG, Nahmgoong H, Oh J, Lee D, Kim DW, Kim JE, Kim YY, Ji Y, Han JS, Kim SM, Sohn JH, Lee WT, Kim SW, Park J, Huh JY, Jo K, Cho JY, Park J, Kim JB. Ubiquitin ligase RNF20 coordinates sequential adipose thermogenesis with brown and beige fat-specific substrates. Nat Commun 2024; 15:940. [PMID: 38296968 PMCID: PMC10831072 DOI: 10.1038/s41467-024-45270-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 01/19/2024] [Indexed: 02/02/2024] Open
Abstract
In mammals, brown adipose tissue (BAT) and inguinal white adipose tissue (iWAT) execute sequential thermogenesis to maintain body temperature during cold stimuli. BAT rapidly generates heat through brown adipocyte activation, and further iWAT gradually stimulates beige fat cell differentiation upon prolonged cold challenges. However, fat depot-specific regulatory mechanisms for thermogenic activation of two fat depots are poorly understood. Here, we demonstrate that E3 ubiquitin ligase RNF20 orchestrates adipose thermogenesis with BAT- and iWAT-specific substrates. Upon cold stimuli, BAT RNF20 is rapidly downregulated, resulting in GABPα protein elevation by controlling protein stability, which stimulates thermogenic gene expression. Accordingly, BAT-specific Rnf20 suppression potentiates BAT thermogenic activity via GABPα upregulation. Moreover, upon prolonged cold stimuli, iWAT RNF20 is gradually upregulated to promote de novo beige adipogenesis. Mechanistically, iWAT RNF20 mediates NCoR1 protein degradation, rather than GABPα, to activate PPARγ. Together, current findings propose fat depot-specific regulatory mechanisms for temporal activation of adipose thermogenesis.
Collapse
Affiliation(s)
- Yong Geun Jeon
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Hahn Nahmgoong
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jiyoung Oh
- Department of Biological Sciences, College of Information and Bioengineering, Ulsan National Institute of Science and Technology, Ulsan, 44919, South Korea
| | - Dabin Lee
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Dong Wook Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Jane Eunsoo Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Ye Young Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Yul Ji
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Ji Seul Han
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Sung Min Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jee Hyung Sohn
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Won Taek Lee
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Sun Won Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jeu Park
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jin Young Huh
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Department of Life Science, Sogang University, Seoul, 04107, South Korea
| | - Kyuri Jo
- Department of Computer Engineering, Chungbuk National University, Cheongju, 28644, South Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Jiyoung Park
- Department of Biological Sciences, College of Information and Bioengineering, Ulsan National Institute of Science and Technology, Ulsan, 44919, South Korea
| | - Jae Bum Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
24
|
Tang S, Yao L, Ruan J, Kang J, Cao Y, Nie X, Lan W, Zhu Z, Han W, Liu Y, Tian J, Seale P, Qin L, Ding C. Single-cell atlas of human infrapatellar fat pad and synovium implicates APOE signaling in osteoarthritis pathology. Sci Transl Med 2024; 16:eadf4590. [PMID: 38266107 DOI: 10.1126/scitranslmed.adf4590] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 09/18/2023] [Indexed: 01/26/2024]
Abstract
The infrapatellar fat pad (IPFP) and synovium play essential roles in maintaining knee joint homeostasis and in the progression of osteoarthritis (OA). The cellular and transcriptional mechanisms regulating the function of these specialized tissues under healthy and diseased conditions are largely unknown. Here, single-cell and single-nuclei RNA sequencing of human IPFP and synovial tissues were performed to elucidate the cellular composition and transcriptional profile. Computational trajectory analysis revealed that dipeptidyl peptidase 4+ mesenchymal cells function as a common progenitor for IPFP adipocytes and synovial lining layer fibroblasts, suggesting that IPFP and synovium represent an integrated tissue unit. OA induced a profibrotic and inflammatory phenotype in mesenchymal lineage cells with biglycan+ intermediate fibroblasts as a major contributor to OA fibrosis. Apolipoprotein E (APOE) signaling from intermediate fibroblasts and macrophages was identified as a critical regulatory factor. Ex vivo incubation of human cartilage with soluble APOE accelerated proteoglycan degeneration. Inhibition of APOE signaling by intra-articular injection of an anti-APOE neutralizing antibody attenuated the progression of collagenase-induced OA in mice, demonstrating a detrimental effect of APOE on cartilage. Our studies provide a framework for designing further therapeutic strategies for OA by describing the cellular and transcriptional landscape of human IPFP and synovium in healthy versus OA joints.
Collapse
Affiliation(s)
- Su'an Tang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
- Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Lutian Yao
- Department of Orthopaedic Surgery, First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jianzhao Ruan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Jingliang Kang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yumei Cao
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Xiaoyu Nie
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Weiren Lan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Weiyu Han
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
- Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yongguang Liu
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Jing Tian
- Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Patrick Seale
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| |
Collapse
|
25
|
Liu W, Liu T, Zhao Q, Ma J, Jiang J, Shi H. Adipose Tissue-Derived Extracellular Vesicles: A Promising Biomarker and Therapeutic Strategy for Metabolic Disorders. Stem Cells Int 2023; 2023:9517826. [PMID: 38169960 PMCID: PMC10761228 DOI: 10.1155/2023/9517826] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/23/2023] [Accepted: 11/25/2023] [Indexed: 01/05/2024] Open
Abstract
Adipose tissue plays an important role in systemic energy metabolism, and its dysfunction can lead to severe metabolic disorders. Various cells in adipose tissue communicate with each other to maintain metabolic homeostasis. Extracellular vesicles (EVs) are recognized as novel medium for remote intercellular communication by transferring various bioactive molecules from parental cells to distant target cells. Increasing evidence suggests that the endocrine functions of adipose tissue and even the metabolic homeostasis are largely affected by different cell-derived EVs, such as insulin signaling, lipolysis, and metabolically triggered inflammation regulations. Here, we provide an overview focused on the role of EVs released by different cell types of adipose tissue in metabolic diseases and their possible molecular mechanisms and highlight the potential applications of EVs as biomarkers and therapeutic targets. Moreover, the current EVs-based therapeutic strategies have also been discussed. This trial is registered with NCT05475418.
Collapse
Affiliation(s)
- Wenhui Liu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Tianyan Liu
- Center of Laboratory Medicine, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
| | - Qingyu Zhao
- Department of Nephrology, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
| | - Junqiu Ma
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
- Center of Laboratory Medicine, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
- Center of Laboratory Medicine, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
| | - Hui Shi
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| |
Collapse
|
26
|
Tong Y, Zuo Z, Li X, Li M, Wang Z, Guo X, Wang X, Sun Y, Chen D, Zhang Z. Protective role of perivascular adipose tissue in the cardiovascular system. Front Endocrinol (Lausanne) 2023; 14:1296778. [PMID: 38155947 PMCID: PMC10753176 DOI: 10.3389/fendo.2023.1296778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/27/2023] [Indexed: 12/30/2023] Open
Abstract
This review provides an overview of the key role played by perivascular adipose tissue (PVAT) in the protection of cardiovascular health. PVAT is a specific type of adipose tissue that wraps around blood vessels and has recently emerged as a critical factor for maintenance of vascular health. Through a profound exploration of existing research, this review sheds light on the intricate structural composition and cellular origins of PVAT, with a particular emphasis on combining its regulatory functions for vascular tone, inflammation, oxidative stress, and endothelial function. The review then delves into the intricate mechanisms by which PVAT exerts its protective effects, including the secretion of diverse adipokines and manipulation of the renin-angiotensin complex. The review further examines the alterations in PVAT function and phenotype observed in several cardiovascular diseases, including atherosclerosis, hypertension, and heart failure. Recognizing the complex interactions of PVAT with the cardiovascular system is critical for pursuing breakthrough therapeutic strategies that can target cardiovascular disease. Therefore, this review aims to augment present understanding of the protective role of PVAT in cardiovascular health, with a special emphasis on elucidating potential mechanisms and paving the way for future research directions in this evolving field.
Collapse
Affiliation(s)
- Yi Tong
- Center for Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zheng Zuo
- Center for Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xinqi Li
- Center for Cardiovascular Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Minghua Li
- Center for Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zhenggui Wang
- Center for Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xiaoxue Guo
- Center for Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xishu Wang
- Center for Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Ying Sun
- Center for Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Dongmei Chen
- Center for Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zhiguo Zhang
- Center for Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
27
|
Lyu QR, Fu K. Tissue-specific Cre driver mice to study vascular diseases. Vascul Pharmacol 2023; 153:107241. [PMID: 37923099 DOI: 10.1016/j.vph.2023.107241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Vascular diseases, including atherosclerosis and abdominal aneurysms, are the primary cause of mortality and morbidity among the elderly worldwide. The life quality of patients is significantly compromised due to inadequate therapeutic approaches and limited drug targets. To expand our comprehension of vascular diseases, gene knockout (KO) mice, especially conditional knockout (cKO) mice, are widely used for investigating gene function and mechanisms of action. The Cre-loxP system is the most common method for generating cKO mice. Numerous Cre driver mice have been established to study the main cell types that compose blood vessels, including endothelial cells, smooth muscle cells, and fibroblasts. Here, we first discuss the characteristics of each layer of the arterial wall. Next, we provide an overview of the representative Cre driver mice utilized for each of the major cell types in the vessel wall and their most recent applications in vascular biology. We then go over Cre toxicity and discuss the practical methods for minimizing Cre interference in experimental outcomes. Finally, we look into the future of tissue-specific Cre drivers by introducing the revolutionary single-cell RNA sequencing and dual recombinase system.
Collapse
Affiliation(s)
- Qing Rex Lyu
- Medical Research Center, Chongqing General Hospital, Chongqing 401147, China; Chongqing Academy of Medical Sciences, Chongqing 401147, China.
| | - Kailong Fu
- Department of Traditional Chinese Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| |
Collapse
|
28
|
You W, Xu Z, Chen W, Yang X, Liu S, Wang L, Tu Y, Zhou Y, Valencak TG, Wang Y, Kuang S, Shan T. Cellular and Transcriptional Dynamics during Brown Adipose Tissue Regeneration under Acute Injury. RESEARCH (WASHINGTON, D.C.) 2023; 6:0268. [PMID: 38434240 PMCID: PMC10907023 DOI: 10.34133/research.0268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 10/18/2023] [Indexed: 03/05/2024]
Abstract
Brown adipose tissue (BAT) is the major site of non-shivering thermogenesis and crucial for systemic metabolism. Under chronic cold exposures and high-fat diet challenges, BAT undergoes robust remodeling to adapt to physiological demands. However, whether and how BAT regenerates after acute injuries are poorly understood. Here, we established a novel BAT injury and regeneration model (BAT-IR) in mice and performed single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq to determine cellular and transcriptomic dynamics during BAT-IR. We further defined distinct fibro-adipogenic and myeloid progenitor populations contributing to BAT regeneration. Cell trajectory and gene expression analyses uncovered the involvement of MAPK, Wnt, and Hedgehog (Hh) signaling pathways in BAT regeneration. We confirmed the role of Hh signaling in BAT development through Myf5Cre-mediated conditional knockout (cKO) of the Sufu gene to activate Hh signaling in BAT and muscle progenitors. Our BAT-IR model therefore provides a paradigm to identify conserved cellular and molecular mechanisms underlying BAT development and remodeling.
Collapse
Affiliation(s)
- Wenjing You
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Ziye Xu
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- Department of Laboratory Medicine, the First Affiliated Hospital, College of Medicine,
Zhejiang University, Hangzhou, China
| | - Wentao Chen
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Xin Yang
- Department of Animal Sciences,
Purdue University, West Lafayette, IN, USA
| | - Shiqi Liu
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Liyi Wang
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Yuang Tu
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Yanbing Zhou
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | | | - Yizhen Wang
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Shihuan Kuang
- Department of Animal Sciences,
Purdue University, West Lafayette, IN, USA
| | - Tizhong Shan
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| |
Collapse
|
29
|
Rao J, Djeffal Y, Chal J, Marchianò F, Wang CH, Al Tanoury Z, Gapon S, Mayeuf-Louchart A, Glass I, Sefton EM, Habermann B, Kardon G, Watt FM, Tseng YH, Pourquié O. Reconstructing human brown fat developmental trajectory in vitro. Dev Cell 2023; 58:2359-2375.e8. [PMID: 37647896 PMCID: PMC10873093 DOI: 10.1016/j.devcel.2023.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 08/23/2022] [Accepted: 08/01/2023] [Indexed: 09/01/2023]
Abstract
Brown adipocytes (BAs) represent a specialized cell type that is able to uncouple nutrient catabolism from ATP generation to dissipate energy as heat. In humans, the brown fat tissue is composed of discrete depots found throughout the neck and trunk region. BAs originate from a precursor common to skeletal muscle, but their developmental trajectory remains poorly understood. Here, we used single-cell RNA sequencing to characterize the development of interscapular brown fat in mice. Our analysis identified a transient stage of BA differentiation characterized by the expression of the transcription factor GATA6. We show that recapitulating the sequence of signaling cues identified in mice can lead to efficient differentiation of BAs in vitro from human pluripotent stem cells. These precursors can in turn be efficiently converted into functional BAs that can respond to signals mimicking adrenergic stimuli by increasing their metabolism, resulting in heat production.
Collapse
Affiliation(s)
- Jyoti Rao
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Yannis Djeffal
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Jerome Chal
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Fabio Marchianò
- Aix-Marseille University, CNRS, IBDM, The Turing Center for Living Systems, 13009 Marseille, France
| | - Chih-Hao Wang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ziad Al Tanoury
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Svetlana Gapon
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | | | - Ian Glass
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Elizabeth M Sefton
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Bianca Habermann
- Aix-Marseille University, CNRS, IBDM, The Turing Center for Living Systems, 13009 Marseille, France
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Fiona M Watt
- King's College London Centre for Stem Cells and Regenerative Medicine, Great Maze Pond, London SE1 9RT, UK
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
30
|
Jun S, Angueira AR, Fein EC, Tan JME, Weller AH, Cheng L, Batmanov K, Ishibashi J, Sakers AP, Stine RR, Seale P. Control of murine brown adipocyte development by GATA6. Dev Cell 2023; 58:2195-2205.e5. [PMID: 37647897 PMCID: PMC10842351 DOI: 10.1016/j.devcel.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 06/07/2023] [Accepted: 08/01/2023] [Indexed: 09/01/2023]
Abstract
Brown adipose tissue (BAT) is a thermogenic organ that protects animals against hypothermia and obesity. BAT derives from the multipotent paraxial mesoderm; however, the identity of embryonic brown fat progenitor cells and regulators of adipogenic commitment are unclear. Here, we performed single-cell gene expression analyses of mesenchymal cells during mouse embryogenesis with a focus on BAT development. We identified cell populations associated with the development of BAT, including Dpp4+ cells that emerge at the onset of adipogenic commitment. Immunostaining and lineage-tracing studies show that Dpp4+ cells constitute the BAT fascia and contribute minorly as adipocyte progenitors. Additionally, we identified the transcription factor GATA6 as a marker of brown adipogenic progenitor cells. Deletion of Gata6 in the brown fat lineage resulted in a striking loss of BAT. Together, these results identify progenitor and transitional cells in the brown adipose lineage and define a crucial role for GATA6 in BAT development.
Collapse
Affiliation(s)
- Seoyoung Jun
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anthony R Angueira
- Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ethan C Fein
- Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Josephine M E Tan
- Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Angela H Weller
- Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lan Cheng
- Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kirill Batmanov
- Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeff Ishibashi
- Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexander P Sakers
- Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rachel R Stine
- Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
31
|
Yang C, Yang X, Harrington A, Potts C, Kaija A, Ryzhova L, Liaw L. Notch Signaling Regulates Mouse Perivascular Adipose Tissue Function via Mitochondrial Pathways. Genes (Basel) 2023; 14:1964. [PMID: 37895313 PMCID: PMC10606454 DOI: 10.3390/genes14101964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Perivascular adipose tissue (PVAT) regulates vascular function by secreting vasoactive substances. In mice, Notch signaling is activated in the PVAT during diet-induced obesity, and leads to the loss of the thermogenic phenotype and adipocyte whitening due to increased lipid accumulation. We used the Adiponectin-Cre (Adipoq-Cre) strain to activate a ligand-independent Notch1 intracellular domain transgene (N1ICD) to drive constitutive Notch signaling in the adipose tissues (N1ICD;Adipoq-Cre). We previously found that constitutive activation of Notch1 signaling in the PVAT phenocopied the effects of diet-induced obesity. To understand the downstream pathways activated by Notch signaling, we performed a proteomic analysis of the PVAT from control versus N1ICD;Adipoq-Cre mice. This comparison identified prominent changes in the protein signatures related to metabolism, adipocyte homeostasis, mitochondrial function, and ferroptosis. PVAT-derived stromal vascular fraction cells were derived from our mouse strains to study the cellular and molecular phenotypes during adipogenic induction. We found that cells with activated Notch signaling displayed decreased mitochondrial respiration despite similar levels of adipogenesis and mitochondrial number. We observed variable regulation of the proteins related to mitochondrial dynamics and ferroptosis, including PHB3, PINK1, pDRP1, and the phospholipid hydroperoxidase GPX4. Mitochondria regulate some forms of ferroptosis, which is a regulated process of cell death driven by lipid peroxidation. Accordingly, we found that Notch activation promoted lipid peroxidation and ferroptosis in PVAT-derived adipocytes. Because the PVAT phenotype is a regulator of vascular reactivity, we tested the effect of Notch activation in PVAT on vasoreactivity using wire myography. The aortae from the N1ICD;Adipoq-Cre mice had increased vasocontraction and decreased vasorelaxation in a PVAT-dependent and age-dependent manner. Our data provide support for the novel concept that increased Notch signaling in the adipose tissue leads to PVAT whitening, impaired mitochondrial function, increased ferroptosis, and loss of a protective vasodilatory signal. Our study advances our understanding of how Notch signaling in adipocytes affects mitochondrial dynamics, which impacts vascular physiology.
Collapse
Affiliation(s)
- Chenhao Yang
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.Y.); (X.Y.); (A.H.); (C.P.); (A.K.); (L.R.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | - Xuehui Yang
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.Y.); (X.Y.); (A.H.); (C.P.); (A.K.); (L.R.)
| | - Anne Harrington
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.Y.); (X.Y.); (A.H.); (C.P.); (A.K.); (L.R.)
| | - Christian Potts
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.Y.); (X.Y.); (A.H.); (C.P.); (A.K.); (L.R.)
| | - Abigail Kaija
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.Y.); (X.Y.); (A.H.); (C.P.); (A.K.); (L.R.)
| | - Larisa Ryzhova
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.Y.); (X.Y.); (A.H.); (C.P.); (A.K.); (L.R.)
| | - Lucy Liaw
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.Y.); (X.Y.); (A.H.); (C.P.); (A.K.); (L.R.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| |
Collapse
|
32
|
Agarwal AK, Tunison K, Vale G, McDonald JG, Li X, Scherer PE, Horton JD, Garg A. Regulated adipose tissue-specific expression of human AGPAT2 in lipodystrophic Agpat2-null mice results in regeneration of adipose tissue. iScience 2023; 26:107806. [PMID: 37752957 PMCID: PMC10518674 DOI: 10.1016/j.isci.2023.107806] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/28/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023] Open
Abstract
Genetic loss of Agpat2 in humans and mice results in congenital generalized lipodystrophy with near-total loss of adipose tissue and predisposition to develop insulin resistance, diabetes mellitus, hepatic steatosis, and hypertriglyceridemia. The mechanism by which Agpat2 deficiency results in loss of adipose tissue remains unknown. We studied this by re-expressing human AGPAT2 (hAGPAT2) in Agpat2-null mice, regulated by doxycycline. In both sexes of Agpat2-null mice, adipose-tissue-specific re-expression of hAGPAT2 resulted in partial regeneration of both white and brown adipose tissue (but only 30%-50% compared with wild-type mice), which had molecular signatures of adipocytes, including leptin secretion. Furthermore, the stromal vascular fraction cells of regenerated adipose depots differentiated ex vivo only with doxycycline, suggesting the essential role of Agpat2 in adipocyte differentiation. Turning off expression of hAGPAT2 in vivo resulted in total loss of regenerated adipose tissue, clear evidence that Agpat2 is essential for adipocyte differentiation in vivo.
Collapse
Affiliation(s)
- Anil K. Agarwal
- Section of Nutrition and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Katie Tunison
- Section of Nutrition and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Goncalo Vale
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeffrey G. McDonald
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xilong Li
- Peter O’Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Philipp E. Scherer
- Touchstone Center for Diabetes Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jay D. Horton
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Abhimanyu Garg
- Section of Nutrition and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
33
|
Thompson JM, Watts SW, Terrian L, Contreras GA, Rockwell C, Rendon CJ, Wabel E, Lockwood L, Bhattacharya S, Nault R. A Cell Atlas of Thoracic Aortic Perivascular Adipose Tissue: a focus on mechanotransducers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.561581. [PMID: 37873456 PMCID: PMC10592719 DOI: 10.1101/2023.10.09.561581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Perivascular adipose tissue (PVAT) is increasingly recognized for its function in mechanotransduction. To examine the cell-specificity of recognized mechanotransducers we used single nuclei RNA sequencing (snRNAseq) of the thoracic aorta PVAT (taPVAT) from male Dahl SS rats compared to subscapular brown adipose tissue (BAT). Approximately 30,000 nuclei from taPVAT and BAT each were characterized by snRNAseq, identifying 8 major cell types expected and one unexpected (nuclei with oligodendrocyte marker genes). Cell-specific differential gene expression analysis between taPVAT and BAT identified up to 511 genes (adipocytes) with many (≥20%) being unique to individual cell types. Piezo1 was the most highly, widely expressed mechanotransducer. Presence of PIEZO1 in the PVAT was confirmed by RNAscope® and IHC; antagonism of PIEZO1 impaired the PVAT's ability to hold tension. Collectively, the cell compositions of taPVAT and BAT are highly similar, and PIEZO1 is likely a mechanotransducer in taPVAT.
Collapse
Affiliation(s)
- Janice M. Thompson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Denotes individuals contributed equally as first authors to this work
| | - Stephanie W. Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Denotes individuals contributed equally as first authors to this work
| | - Leah Terrian
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Denotes individuals contributed equally as first authors to this work
| | - G. Andres Contreras
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - Cheryl Rockwell
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - C. Javier Rendon
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - Emma Wabel
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Lizabeth Lockwood
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Sudin Bhattacharya
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Rance Nault
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Denotes individuals contributed equally as first authors to this work
- Denotes lead contact
| |
Collapse
|
34
|
Abstract
Adipose tissue exhibits a remarkable capacity to expand, contract, and remodel in response to changes in physiological and environmental conditions. Here, we describe recent advances in our understanding of how functionally distinct tissue-resident mesenchymal stromal cell subpopulations orchestrate several aspects of physiological and pathophysiological adipose tissue remodeling, with a particular focus on the adaptations that occur in response to changes in energy surplus and environmental temperature. The study of adipose tissue remodeling provides a vehicle to understand the functional diversity of stromal cells and offers a lens through which several generalizable aspects of tissue reorganization can be readily observed.
Collapse
Affiliation(s)
- Jessica Cannavino
- Department of Medicine, Division of Endocrinology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina 27701, USA
| | - Rana K Gupta
- Department of Medicine, Division of Endocrinology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina 27701, USA
| |
Collapse
|
35
|
Zhang K, Zhang J, Kan C, Tian H, Ma Y, Huang N, Han F, Hou N, Sun X. Role of dysfunctional peri-organ adipose tissue in metabolic disease. Biochimie 2023; 212:12-20. [PMID: 37019205 DOI: 10.1016/j.biochi.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023]
Abstract
Metabolic disease is a complex disorder defined by a group with interrelated factors. There is growing evidence that obesity can lead to a variety of metabolic diseases, including diabetes and cardiovascular disease. Excessive adipose tissue (AT) deposition and ectopic accumulation can lead to increased peri-organ AT thickness. Dysregulation of peri-organ (perivascular, perirenal, and epicardial) AT is strongly associated with metabolic disease and its complications. The mechanisms include secretion of cytokines, activation of immunocytes, infiltration of inflammatory cells, involvement of stromal cells, and abnormal miRNA expression. This review discusses the associations and mechanisms by which various types of peri-organ AT affect metabolic diseases while addressing it as a potential future treatment strategy.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hongzhan Tian
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yanhui Ma
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Na Huang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.
| |
Collapse
|
36
|
Yuan Y, Shi Z, Xiong S, Hu R, Song Q, Song Z, Ong SG, Jiang Y. Differential roles of insulin receptor in adipocyte progenitor cells in mice. Mol Cell Endocrinol 2023; 573:111968. [PMID: 37244600 PMCID: PMC10846871 DOI: 10.1016/j.mce.2023.111968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/29/2023]
Abstract
The development of white adipose tissue (WAT) occurs during distinct embryonic and postnatal stages, and it is subsequently maintained throughout life. However, the specific mediators and mechanisms responsible for WAT development during different phases remain unclear. In this study, we investigate the role of the insulin receptor (IR) in regulating adipogenesis and adipocyte function within adipocyte progenitor cells (APCs) during WAT development and homeostasis. We use two in vivo adipose lineage tracking and deletion systems to delete IR either in embryonic APCs or adult APCs, respectively, to explore the specific requirements of IR during WAT development and WAT homeostasis in mice. Our data suggest that IR expression in APCs may not be essential for adult adipocyte differentiation but appears to be crucial for adipose tissue development. We reveal a surprising divergent role of IR in APCs during WAT development and homeostasis.
Collapse
Affiliation(s)
- Yexian Yuan
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Zuoxiao Shi
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, 60612, USA; Department of Pharmaceutical Sciences, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shaolei Xiong
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ruoci Hu
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, 60612, USA; Department of Pharmaceutical Sciences, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Qing Song
- Department of Kinesiology and Nutrition, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sang-Ging Ong
- Department of Pharmacology and Regenerative Medicine, College of Medicine, The University of Illinois at Chicago, Illinois, 60612, USA; Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, Illinois, 60612, USA
| | - Yuwei Jiang
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, 60612, USA; Department of Pharmaceutical Sciences, The University of Illinois at Chicago, Chicago, IL, 60612, USA; Division of Endocrinology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
37
|
Sparks L, Whytock K, Divoux A, Sun Y, Pino M, Yu G, Smith S, Walsh M. A single nuclei atlas of aging human abdominal subcutaneous white adipose tissue. RESEARCH SQUARE 2023:rs.3.rs-3097605. [PMID: 37503028 PMCID: PMC10371078 DOI: 10.21203/rs.3.rs-3097605/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
White adipose tissue (WAT) is a robust energy storage and endocrine organ critical for maintaining metabolic health as we age. Our aim was to identify cell-specific transcriptional aberrations that occur in WAT with aging. We leveraged full-length snRNA-Seq to characterize the cellular landscape of human subcutaneous WAT in a prospective cohort of 10 Younger (≤ 30 years) and 10 Older individuals (≥ 65 years) balanced for sex and body mass index (BMI). We highlight that aging WAT is associated with adipocyte hypertrophy, increased proportions of resident macrophages (M2), an upregulated innate immune response and senescence profiles in specific adipocyte populations, highlighting CXCL14 as a biomarker of this process. We also identify novel markers of pre-adipocytes and track their expression levels through pre-adipocyte differentiation. We propose that aging WAT is associated with low-grade inflammation that is managed by a foundation of innate immunity to preserve the metabolic health of the WAT.
Collapse
Affiliation(s)
| | | | | | - Yifei Sun
- Icahn School of Medicine at Mount Sinai
| | - Maria Pino
- Translational Research Institute, AdventHealth
| | - Gongxin Yu
- Translational Research Institute, AdventHealth
| | | | | |
Collapse
|
38
|
Wang C, Wang X, Hu W. Molecular and cellular regulation of thermogenic fat. Front Endocrinol (Lausanne) 2023; 14:1215772. [PMID: 37465124 PMCID: PMC10351381 DOI: 10.3389/fendo.2023.1215772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/14/2023] [Indexed: 07/20/2023] Open
Abstract
Thermogenic fat, consisting of brown and beige adipocytes, dissipates energy in the form of heat, in contrast to the characteristics of white adipocytes that store energy. Increasing energy expenditure by activating brown adipocytes or inducing beige adipocytes is a potential therapeutic strategy for treating obesity and type 2 diabetes. Thus, a better understanding of the underlying mechanisms of thermogenesis provides novel therapeutic interventions for metabolic diseases. In this review, we summarize the recent advances in the molecular regulation of thermogenesis, focusing on transcription factors, epigenetic regulators, metabolites, and non-coding RNAs. We further discuss the intercellular and inter-organ crosstalk that regulate thermogenesis, considering the heterogeneity and complex tissue microenvironment of thermogenic fat.
Collapse
Affiliation(s)
- Cuihua Wang
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangdong, China
| | - Xianju Wang
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Wenxiang Hu
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
39
|
Klarin D, Devineni P, Sendamarai AK, Angueira AR, Graham SE, Shen YH, Levin MG, Pirruccello JP, Surakka I, Karnam PR, Roychowdhury T, Li Y, Wang M, Aragam KG, Paruchuri K, Zuber V, Shakt GE, Tsao NL, Judy RL, Vy HMT, Verma SS, Rader DJ, Do R, Bavaria JE, Nadkarni GN, Ritchie MD, Burgess S, Guo DC, Ellinor PT, LeMaire SA, Milewicz DM, Willer CJ, Natarajan P, Tsao PS, Pyarajan S, Damrauer SM. Genome-wide association study of thoracic aortic aneurysm and dissection in the Million Veteran Program. Nat Genet 2023; 55:1106-1115. [PMID: 37308786 PMCID: PMC10335930 DOI: 10.1038/s41588-023-01420-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/05/2023] [Indexed: 06/14/2023]
Abstract
The current understanding of the genetic determinants of thoracic aortic aneurysms and dissections (TAAD) has largely been informed through studies of rare, Mendelian forms of disease. Here, we conducted a genome-wide association study (GWAS) of TAAD, testing ~25 million DNA sequence variants in 8,626 participants with and 453,043 participants without TAAD in the Million Veteran Program, with replication in an independent sample of 4,459 individuals with and 512,463 without TAAD from six cohorts. We identified 21 TAAD risk loci, 17 of which have not been previously reported. We leverage multiple downstream analytic methods to identify causal TAAD risk genes and cell types and provide human genetic evidence that TAAD is a non-atherosclerotic aortic disorder distinct from other forms of vascular disease. Our results demonstrate that the genetic architecture of TAAD mirrors that of other complex traits and that it is not solely inherited through protein-altering variants of large effect size.
Collapse
Affiliation(s)
- Derek Klarin
- Veterans Affairs (VA) Palo Alto Healthcare System, Palo Alto, CA, USA.
- Department of Surgery, Stanford University School of Medicine, Palo Alto, CA, USA.
| | - Poornima Devineni
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
| | - Anoop K Sendamarai
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Anthony R Angueira
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah E Graham
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, USA
| | - Michael G Levin
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Medicine, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - James P Pirruccello
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Ida Surakka
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Purushotham R Karnam
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
| | - Tanmoy Roychowdhury
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Minxian Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Krishna G Aragam
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kaavya Paruchuri
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Verena Zuber
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
| | - Gabrielle E Shakt
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Noah L Tsao
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Renae L Judy
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Ha My T Vy
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shefali S Verma
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel J Rader
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ron Do
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph E Bavaria
- Division of Cardiovascular Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Marylyn D Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Biomedical Informatics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Stephen Burgess
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Dong-Chuan Guo
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Patrick T Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Dianna M Milewicz
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Cristen J Willer
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Pradeep Natarajan
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Philip S Tsao
- Veterans Affairs (VA) Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Saiju Pyarajan
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Scott M Damrauer
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA.
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Vallecillo-García P, Orgeur M, Comai G, Poehle-Kronawitter S, Fischer C, Gloger M, Dumas CE, Giesecke-Thiel C, Sauer S, Tajbakhsh S, Höpken UE, Stricker S. A local subset of mesenchymal cells expressing the transcription factor Osr1 orchestrates lymph node initiation. Immunity 2023; 56:1204-1219.e8. [PMID: 37160119 DOI: 10.1016/j.immuni.2023.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 12/05/2022] [Accepted: 04/13/2023] [Indexed: 05/11/2023]
Abstract
During development, lymph node (LN) initiation is coordinated by lymphoid tissue organizer (LTo) cells that attract lymphoid tissue inducer (LTi) cells at strategic positions within the embryo. The identity and function of LTo cells during the initial attraction of LTi cells remain poorly understood. Using lineage tracing, we demonstrated that a subset of Osr1-expressing cells was mesenchymal LTo progenitors. By investigating the heterogeneity of Osr1+ cells, we uncovered distinct mesenchymal LTo signatures at diverse anatomical locations, identifying a common progenitor of mesenchymal LTos and LN-associated adipose tissue. Osr1 was essential for LN initiation, driving the commitment of mesenchymal LTo cells independent of neural retinoic acid, and for LN-associated lymphatic vasculature assembly. The combined action of chemokines CXCL13 and CCL21 was required for LN initiation. Our results redefine the role and identity of mesenchymal organizer cells and unify current views by proposing a model of cooperative cell function in LN initiation.
Collapse
Affiliation(s)
| | - Mickael Orgeur
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, 75015 Paris, France
| | - Glenda Comai
- Institut Pasteur, Stem Cells & Development Unit, CNRS UMR 3738, Paris, France
| | | | - Cornelius Fischer
- Core Facility Genomics, Berlin Institute of Health at Charité, 10178 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115, Berlin, Germany
| | - Marleen Gloger
- Max Delbrück Center for Molecular Medicine, Department of Translational Tumor Immunology, 13125 Berlin, Germany; Uppsala University, Immunology Genetics and Pathology, 75237 Uppsala, Sweden
| | - Camille E Dumas
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | | | - Sascha Sauer
- Core Facility Genomics, Berlin Institute of Health at Charité, 10178 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115, Berlin, Germany
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells & Development Unit, CNRS UMR 3738, Paris, France
| | - Uta E Höpken
- Max Delbrück Center for Molecular Medicine, Department of Microenvironmental Regulation in Autoimmunity and Cancer, 13125 Berlin, Germany
| | - Sigmar Stricker
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.
| |
Collapse
|
41
|
Abstract
The circadian clock plays an essential role in coordinating feeding and metabolic rhythms with the light/dark cycle. Disruption of clocks is associated with increased adiposity and metabolic disorders, whereas aligning feeding time with cell-autonomous rhythms in metabolism improves health. Here, we provide a comprehensive overview of recent literature in adipose tissue biology as well as our understanding of molecular mechanisms underlying the circadian regulation of transcription, metabolism, and inflammation in adipose tissue. We highlight recent efforts to uncover the mechanistic links between clocks and adipocyte metabolism, as well as its application to dietary and behavioral interventions to improve health and mitigate obesity.
Collapse
Affiliation(s)
- Chelsea Hepler
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| |
Collapse
|
42
|
Adachi Y, Ueda K, Takimoto E. Perivascular adipose tissue in vascular pathologies-a novel therapeutic target for atherosclerotic disease? Front Cardiovasc Med 2023; 10:1151717. [PMID: 37304960 PMCID: PMC10250715 DOI: 10.3389/fcvm.2023.1151717] [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: 01/26/2023] [Accepted: 04/28/2023] [Indexed: 06/13/2023] Open
Abstract
Most blood vessels are surrounded by adipose tissues known as perivascular adipose tissue (PVAT). Emerging experimental data have implicated the potential involvement of PVAT in the pathogenesis of cardiovascular disease: PVAT might be a source of inflammatory mediators under pathological conditions such as metabolic disorders, chronic inflammation, and aging, leading to vascular pathologies, while having vasculo-protective roles in a healthy state. PVAT has been also gaining attention in human disease conditions. Recent integrative omics approaches have greatly enhanced our understanding of the molecular mechanisms underlying the diverse functions of PVAT. This review summarizes recent progress in PVAT research and discusses the potential of PVAT as a target for the treatment of atherosclerosis.
Collapse
|
43
|
Shi Z, Xiong S, Hu R, Wang Z, Park J, Qian Y, Wang J, Bhalla P, Velupally N, Song Q, Song Z, Layden BT, Jiang Y. The Notch-Pdgfrβ axis suppresses brown adipocyte progenitor differentiation in early postnatal mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.541839. [PMID: 37293108 PMCID: PMC10245810 DOI: 10.1101/2023.05.24.541839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
De novo brown adipogenesis holds potential in combating the epidemics of obesity and diabetes. However, the identity of brown adipocyte progenitor cells (APCs) and their regulation have not been extensively studied. Here through in vivo lineage tracing, we observed that PDGFRβ+ pericytes give rise to developmental brown adipocytes, but not to those in adult homeostasis. In contrast, TBX18+ pericytes contribute to brown adipogenesis throughout both developmental and adult stages, though in a depot-specific manner. Mechanistically, Notch inhibition in PDGFRβ+ pericytes promotes brown adipogenesis through the downregulation of PDGFRβ. Furthermore, inhibition of Notch signaling in PDGFRβ+ pericytes mitigates HFHS (high-fat, high-sucrose) induced glucose and metabolic impairment in both developmental and adult stages. Collectively, these findings show that the Notch/PDGFRβ axis negatively regulates developmental brown adipogenesis, and its repression promotes brown adipose tissue expansion and improves metabolic health. Highlights PDGFRβ+ pericytes act as an essential developmental brown APC.TBX18+ pericytes contribute to brown adipogenesis in a depot-specific manner.Inhibiting Notch-Pdgfrβ axis promotes brown APC adipogenesis.Enhanced postnatal brown adipogenesis improves metabolic health in adult stage.
Collapse
|
44
|
Takashima Y, Yamamoto S, Okuno N, Hamashima T, Dang ST, Tran ND, Okita N, Miwa F, Dang TC, Matsuo M, Takao K, Fujimori T, Mori H, Tobe K, Noguchi M, Sasahara M. PDGF receptor signal mediates the contribution of Nestin-positive cell lineage to subcutaneous fat development. Biochem Biophys Res Commun 2023; 658:27-35. [PMID: 37018886 DOI: 10.1016/j.bbrc.2023.03.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 04/05/2023]
Abstract
The beiging of white adipose tissue (WAT) is expected to improve systemic metabolic conditions; however, the regulation and developmental origin of this process remain insufficiently understood. In the present study, the implication of platelet-derived growth factor receptor alpha (PDGFRα) was examined in the beiging of inguinal WAT (ingWAT) of neonatal mice. Using in vivo Nestin expressing cell (Nestin+) lineage tracing and deletion mouse models, we found that, in the mice with Pdgfra gene inactivation in Nestin+ lineage (N-PRα-KO mice), the growth of inguinal WAT (ingWAT) was suppressed during neonatal periods as compared with control wild-type mice. In the ingWAT of N-PRα-KO mice, the beige adipocytes appeared earlier that were accompanied by the increased expressions of both adipogenic and beiging markers compared to control wild-type mice. In the perivascular adipocyte progenitor cell (APC) niche of ingWAT, many PDGFRα+ cells of Nestin+ lineage were recruited in Pdgfra-preserving control mice, but were largely decreased in N-PRα-KO mice. This PDGFRα+ cell depletion was replenished by PDGFRα+ cells of non-Nestin+ lineage, unexpectedly resulting in an increase of total PDGFRα+ cell number in APC niche of N-PRα-KO mice over that of control mice. These represented a potent homeostatic control of PDGFRα+ cells between Nestin+ and non-Nestin+ lineages that was accompanied by the active adipogenesis and beiging as well as small WAT depot. This highly plastic nature of PDGFRα+ cells in APC niche may contribute to the WAT remodeling for the therapeutic purpose against metabolic diseases.
Collapse
|
45
|
Ahmed A, Bibi A, Valoti M, Fusi F. Perivascular Adipose Tissue and Vascular Smooth Muscle Tone: Friends or Foes? Cells 2023; 12:cells12081196. [PMID: 37190105 DOI: 10.3390/cells12081196] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/09/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023] Open
Abstract
Perivascular adipose tissue (PVAT) is a specialized type of adipose tissue that surrounds most mammalian blood vessels. PVAT is a metabolically active, endocrine organ capable of regulating blood vessel tone, endothelium function, vascular smooth muscle cell growth and proliferation, and contributing critically to cardiovascular disease onset and progression. In the context of vascular tone regulation, under physiological conditions, PVAT exerts a potent anticontractile effect by releasing a plethora of vasoactive substances, including NO, H2S, H2O2, prostacyclin, palmitic acid methyl ester, angiotensin 1-7, adiponectin, leptin, and omentin. However, under certain pathophysiological conditions, PVAT exerts pro-contractile effects by decreasing the production of anticontractile and increasing that of pro-contractile factors, including superoxide anion, angiotensin II, catecholamines, prostaglandins, chemerin, resistin, and visfatin. The present review discusses the regulatory effect of PVAT on vascular tone and the factors involved. In this scenario, dissecting the precise role of PVAT is a prerequisite to the development of PVAT-targeted therapies.
Collapse
Affiliation(s)
- Amer Ahmed
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Aasia Bibi
- Nanotechnology Institute, CNR-NANOTEC, Via Monteroni, 73100 Lecce, Italy
| | - Massimo Valoti
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Fabio Fusi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
46
|
An integrated single cell and spatial transcriptomic map of human white adipose tissue. Nat Commun 2023; 14:1438. [PMID: 36922516 PMCID: PMC10017705 DOI: 10.1038/s41467-023-36983-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
To date, single-cell studies of human white adipose tissue (WAT) have been based on small cohort sizes and no cellular consensus nomenclature exists. Herein, we performed a comprehensive meta-analysis of publicly available and newly generated single-cell, single-nucleus, and spatial transcriptomic results from human subcutaneous, omental, and perivascular WAT. Our high-resolution map is built on data from ten studies and allowed us to robustly identify >60 subpopulations of adipocytes, fibroblast and adipogenic progenitors, vascular, and immune cells. Using these results, we deconvolved spatial and bulk transcriptomic data from nine additional cohorts to provide spatial and clinical dimensions to the map. This identified cell-cell interactions as well as relationships between specific cell subtypes and insulin resistance, dyslipidemia, adipocyte volume, and lipolysis upon long-term weight changes. Altogether, our meta-map provides a rich resource defining the cellular and microarchitectural landscape of human WAT and describes the associations between specific cell types and metabolic states.
Collapse
|
47
|
Maniyadath B, Zhang Q, Gupta RK, Mandrup S. Adipose tissue at single-cell resolution. Cell Metab 2023; 35:386-413. [PMID: 36889280 PMCID: PMC10027403 DOI: 10.1016/j.cmet.2023.02.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 03/09/2023]
Abstract
Adipose tissue exhibits remarkable plasticity with capacity to change in size and cellular composition under physiological and pathophysiological conditions. The emergence of single-cell transcriptomics has rapidly transformed our understanding of the diverse array of cell types and cell states residing in adipose tissues and has provided insight into how transcriptional changes in individual cell types contribute to tissue plasticity. Here, we present a comprehensive overview of the cellular atlas of adipose tissues focusing on the biological insight gained from single-cell and single-nuclei transcriptomics of murine and human adipose tissues. We also offer our perspective on the exciting opportunities for mapping cellular transitions and crosstalk, which have been made possible by single-cell technologies.
Collapse
Affiliation(s)
- Babukrishna Maniyadath
- Center for Functional Genomics and Tissue Plasticity, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Qianbin Zhang
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rana K Gupta
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark.
| |
Collapse
|
48
|
Abstract
Brown adipose tissue (BAT) displays the unique capacity to generate heat through uncoupled oxidative phosphorylation that makes it a very attractive therapeutic target for cardiometabolic diseases. Here, we review BAT cellular metabolism, its regulation by the central nervous and endocrine systems and circulating metabolites, the plausible roles of this tissue in human thermoregulation, energy balance, and cardiometabolic disorders, and the current knowledge on its pharmacological stimulation in humans. The current definition and measurement of BAT in human studies relies almost exclusively on BAT glucose uptake from positron emission tomography with 18F-fluorodeoxiglucose, which can be dissociated from BAT thermogenic activity, as for example in insulin-resistant states. The most important energy substrate for BAT thermogenesis is its intracellular fatty acid content mobilized from sympathetic stimulation of intracellular triglyceride lipolysis. This lipolytic BAT response is intertwined with that of white adipose (WAT) and other metabolic tissues, and cannot be independently stimulated with the drugs tested thus far. BAT is an interesting and biologically plausible target that has yet to be fully and selectively activated to increase the body's thermogenic response and shift energy balance. The field of human BAT research is in need of methods able to directly, specifically, and reliably measure BAT thermogenic capacity while also tracking the related thermogenic responses in WAT and other tissues. Until this is achieved, uncertainty will remain about the role played by this fascinating tissue in human cardiometabolic diseases.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | | | - Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec City, Quebec, G1V 4G5, Canada
| |
Collapse
|
49
|
Mengozzi A, Costantino S, Mongelli A, Mohammed SA, Gorica E, Delfine V, Masi S, Virdis A, Ruschitzka F, Paneni F. Epigenetic Signatures in Arterial Hypertension: Focus on the Microvasculature. Int J Mol Sci 2023; 24:ijms24054854. [PMID: 36902291 PMCID: PMC10003673 DOI: 10.3390/ijms24054854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/25/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Systemic arterial hypertension (AH) is a multifaceted disease characterized by accelerated vascular aging and high cardiometabolic morbidity and mortality. Despite extensive work in the field, the pathogenesis of AH is still incompletely understood, and its treatment remains challenging. Recent evidence has shown a deep involvement of epigenetic signals in the regulation of transcriptional programs underpinning maladaptive vascular remodeling, sympathetic activation and cardiometabolic alterations, all factors predisposing to AH. After occurring, these epigenetic changes have a long-lasting effect on gene dysregulation and do not seem to be reversible upon intensive treatment or the control of cardiovascular risk factors. Among the factors involved in arterial hypertension, microvascular dysfunction plays a central role. This review will focus on the emerging role of epigenetic changes in hypertensive-related microvascular disease, including the different cell types and tissues (endothelial cells, vascular smooth muscle cells and perivascular adipose tissue) as well as the involvement of mechanical/hemodynamic factors, namely, shear stress.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Health Science Interdisciplinary Center, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Alessia Mongelli
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Shafeeq A. Mohammed
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Valentina Delfine
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, 8091 Zurich, Switzerland
- Correspondence: or francesco.paneni@uzh; Tel.: +41-44-6355096
| |
Collapse
|
50
|
Takeda Y, Harada Y, Yoshikawa T, Dai P. Mitochondrial Energy Metabolism in the Regulation of Thermogenic Brown Fats and Human Metabolic Diseases. Int J Mol Sci 2023; 24:ijms24021352. [PMID: 36674862 PMCID: PMC9861294 DOI: 10.3390/ijms24021352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Brown fats specialize in thermogenesis by increasing the utilization of circulating blood glucose and fatty acids. Emerging evidence suggests that brown adipose tissue (BAT) prevents the incidence of obesity-associated metabolic diseases and several types of cancers in humans. Mitochondrial energy metabolism in brown/beige adipocytes regulates both uncoupling protein 1 (UCP1)-dependent and -independent thermogenesis for cold adaptation and the utilization of excess nutrients and energy. Many studies on the quantification of human BAT indicate that mass and activity are inversely correlated with the body mass index (BMI) and visceral adiposity. Repression is caused by obesity-associated positive and negative factors that control adipocyte browning, de novo adipogenesis, mitochondrial energy metabolism, UCP1 expression and activity, and noradrenergic response. Systemic and local factors whose levels vary between lean and obese conditions include growth factors, inflammatory cytokines, neurotransmitters, and metal ions such as selenium and iron. Modulation of obesity-associated repression in human brown fats is a promising strategy to counteract obesity and related metabolic diseases through the activation of thermogenic capacity. In this review, we highlight recent advances in mitochondrial metabolism, thermogenic regulation of brown fats, and human metabolic diseases.
Collapse
Affiliation(s)
- Yukimasa Takeda
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Correspondence: (Y.T.); (P.D.); Tel.: +81-75-251-5444 (Y.T.); +81-75-251-5135 (P.D.)
| | - Yoshinori Harada
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshikazu Yoshikawa
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Louis Pasteur Center for Medical Research, 103-5 Tanaka-Monzen-cho, Sakyo-ku, Kyoto 606-8225, Japan
| | - Ping Dai
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Correspondence: (Y.T.); (P.D.); Tel.: +81-75-251-5444 (Y.T.); +81-75-251-5135 (P.D.)
| |
Collapse
|