1
|
Wang H, Qin Y, Niu J, Chen H, Lu X, Wang R, Han J. Evolving perspectives on evaluating obesity: from traditional methods to cutting-edge techniques. Ann Med 2025; 57:2472856. [PMID: 40077889 PMCID: PMC11912248 DOI: 10.1080/07853890.2025.2472856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Objective: This review examines the evolution of obesity evaluation methods, from traditional anthropometric indices to advanced imaging techniques, focusing on their clinical utility, limitations, and potential for personalized assessment of visceral adiposity and associated metabolic risks. Methods: A comprehensive analysis of existing literature was conducted, encompassing anthropometric indices (BMI, WC, WHR, WHtR, NC), lipid-related metrics (LAP, VAI, CVAI, mBMI), and imaging technologies (3D scanning, BIA, ultrasound, DXA, CT, MRI). The study highlights the biological roles of white, brown, and beige adipocytes, emphasizing visceral adipose tissue (VAT) as a critical mediator of metabolic diseases. Conclusion: Although BMI and other anthropometric measurements are still included in the guidelines, indicators that incorporate lipid metabolism information can more accurately reflect the relationship between metabolic diseases and visceral obesity. At the same time, the use of more modern medical equipment, such as ultrasound, X-rays, and CT scans, allows for a more intuitive assessment of the extent of visceral obesity.
Collapse
Affiliation(s)
- Heyue Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaxin Qin
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinzhu Niu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haowen Chen
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinda Lu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianli Han
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
2
|
Sugino T, Taguchi K, Yanase T, Unno R, Ando R, Yasui T. Brown adipose tissue detection using positron emission tomography could help reduce urolithiasis risk. Urolithiasis 2025; 53:64. [PMID: 40167829 DOI: 10.1007/s00240-025-01739-8] [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: 12/01/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
Urolithiasis is associated with metabolic syndrome, and reactivation of brown adipose tissue (BAT) may improve metabolic syndrome. In this study, we aimed to evaluate the association of BAT, as detected using positron emission tomography-computed tomography (PET-CT), with urolithiasis in humans. This single center retrospective cohort study involved patients who underwent PET-CT for cancer screening as part of a medical checkup between January 2006 and December 2020. We obtained data on participant demographics, presence of urolithiasis, and metabolic factors (such as obesity, hypertension, diabetes, and hyperlipidemia) from 182 medical records. BAT data and calcification of the abdominal aorta (CAA) rate, determined using abdominal CT, were also recorded. Any association between urolithiasis and other factors was evaluated using logistic regression analysis. Body mass index was higher in participants with BAT than in those without BAT (26.92 vs. 22.86 kg/m2, p = 0.001). Participants with BAT had less urolithiasis and a lower CAA rate than those without BAT (10% vs. 37%, p = 0.031 and 50% vs. 79.6%, p = 0.008, respectively). Metabolic factors between the groups were similar (45.0% vs. 48.8%, p = 0.936). Furthermore, BAT and age were associated with a decreased odds ratio (OR) for urolithiasis (OR = 0.186, p = 0.037 and OR = 0.959, p = 0.02, respectively). We demonstrated that high BAT content is associated with a low risk of urolithiasis and CAA. Our findings may contribute to the development of novel preventive methods for urolithiasis.
Collapse
Affiliation(s)
- Teruaki Sugino
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Urology, Nagoya City University East Medical Center, Nagoya, Japan
| | - Kazumi Taguchi
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | - Takahiro Yanase
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Rei Unno
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ryosuke Ando
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahiro Yasui
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
3
|
Gao Z, Yu Y, Eckel‐Mahan K, Kolonin MG. Caloric Restriction and Telomere Preservation in TERT Knockout Adipocyte Progenitors Does Not Rescue Mice From Metabolic Dysfunction due to a TERT Function in Adipocyte Mitochondria. Aging Cell 2025; 24:e14499. [PMID: 39932851 PMCID: PMC11896407 DOI: 10.1111/acel.14499] [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: 11/04/2024] [Revised: 12/20/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
Inactivation of telomerase (TERT) in adipocyte progenitor cells (APC) expedites telomere attrition, and the onset of diabetes in mice fed high-fat diet (HFD), which promotes APC over-proliferation and replicative senescence. Here, we show that time-restricted feeding or caloric restriction in the postnatal development of mice subsequently subjected to HFD prevents telomere attrition but not glucose intolerance. This metabolic effect of dietary intervention was not observed for mice with TERT KO in endothelial or myeloid cells. To characterize the telomere-independent effects of TERT in the APC lineage, we analyzed mice with TERT knockout in mature adipocytes (AD-TERT-KO), which do not proliferate and avoid telomere attrition. Analysis of adipocytes from AD-TERT-KO mice indicated reliance on glycolysis and decreased mitochondrial oxidative metabolism. We show that AD-TERT-KO mice have reduced cold tolerance and metabolism abnormality indicating a defect in adaptive thermogenesis, characteristic of aging. Conversely, ectopic TERT expression in brown adipocytes-induced mitochondrial oxidation and thermogenic gene expression. We conclude that TERT plays an important non-canonical function in the mitochondria of adipocytes.
Collapse
Affiliation(s)
- Zhanguo Gao
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Kristin Eckel‐Mahan
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| |
Collapse
|
4
|
Wang FD, Ding Y, Zhou JH, Zhou E, Zhang TT, Fan YQ, He Q, Zhang ZQ, Mao CY, Zhang JF, Zhou J. Gamma-aminobutyric acid enhances miR-21-5p loading into adipose-derived stem cell extracellular vesicles to alleviate myocardial ischemia-reperfusion injury via TXNIP regulation. World J Stem Cells 2024; 16:873-895. [PMID: 39493825 PMCID: PMC11525649 DOI: 10.4252/wjsc.v16.i10.873] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/21/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury (MIRI) poses a prevalent challenge in current reperfusion therapies, with an absence of efficacious interventions to address the underlying causes. AIM To investigate whether the extracellular vesicles (EVs) secreted by adipose mesenchymal stem cells (ADSCs) derived from subcutaneous inguinal adipose tissue (IAT) under γ-aminobutyric acid (GABA) induction (GABA-EVsIAT) demonstrate a more pronounced inhibitory effect on mitochondrial oxidative stress and elucidate the underlying mechanisms. METHODS We investigated the potential protective effects of EVs derived from mouse ADSCs pretreated with GABA. We assessed cardiomyocyte injury using terminal deoxynucleotidyl transferase dUTP nick end-labeling and Annexin V/propidium iodide assays. The integrity of cardiomyocyte mitochondria morphology was assessed using electron microscopy across various intervention backgrounds. To explore the functional RNA diversity between EVsIAT and GABA-EVsIAT, we employed microRNA (miR) sequencing. Through a dual-luciferase reporter assay, we confirmed the molecular mechanism by which EVs mediate thioredoxin-interacting protein (TXNIP). Western blotting and immunofluorescence were conducted to determine how TXNIP is involved in mediation of oxidative stress and mitochondrial dysfunction. RESULTS Our study demonstrates that, under the influence of GABA, ADSCs exhibit an increased capacity to encapsulate a higher abundance of miR-21-5p within EVs. Consequently, this leads to a more pronounced inhibitory effect on mitochondrial oxidative stress compared to EVs from ADSCs without GABA intervention, ultimately resulting in myocardial protection. On a molecular mechanism level, EVs regulate the expression of TXNIP and mitigating excessive oxidative stress in mitochondria during MIRI process to rescue cardiomyocytes. CONCLUSION Administration of GABA leads to the specific loading of miR-21-5p into EVs by ADSCs, thereby regulating the expression of TXNIP. The EVs derived from ADSCs treated with GABA effectively ameliorates mitochondrial oxidative stress and mitigates cardiomyocytes damage in the pathological process of MIRI.
Collapse
Affiliation(s)
- Feng-Dan Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yi Ding
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jian-Hong Zhou
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - En Zhou
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Tian-Tian Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yu-Qi Fan
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qing He
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zong-Qi Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Cheng-Yu Mao
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jun-Feng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jing Zhou
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
5
|
Xiang K, Pan J, Yu J, Xiao L, Sun SK, Cheng R. A hemicyanine-based near-infrared fluorescent probe with large Stokes shift for non-invasive bioimaging of brown adipose tissue. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:5272-5279. [PMID: 39016035 DOI: 10.1039/d4ay00658e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Brown adipose tissue (BAT), characterized by the presence of numerous mitochondria, plays a key role in metabolism and energy expenditure. Accurately reporting the presence and activation of BAT is beneficial to study obesity, diabetes, and other metabolic disorders. Near-infrared (NIR) fluorescence imaging has the advantages of high sensitivity, non-radioactivity, and simple operation. However, most NIR probes for BAT imaging exhibit small Stokes shifts, which may lead to self-quenching, reducing the signal-to-noise ratio, and introducing cross-talk. Herein, we rationally designed and synthesized a series of hemicyanine-based NIR fluorescent probes HCYBAT-1-3. Among them, HCYBAT-1 demonstrated favorable properties such as near-infrared emission (776 nm), large Stokes shift (139 nm), good biocompatibility and specific mitochondrial targeting (Pearson's colocalization coefficient of 0.87). Meanwhile, HCYBAT-1 was successfully employed to differentiate BAT from white adipose tissue (WAT). Quantitative analysis of NIR fluorescent images showed that HCYBAT-1 could be used for real-time monitoring of BAT activation in mice stimulated by norepinephrine (NE) and cold exposure. Overall, probe HCYBAT-1 showcased its efficacy in non-invasive evaluation of BAT metabolism in vivo with high selectivity and sensitivity.
Collapse
Affiliation(s)
- Ke Xiang
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China.
| | - Jinbin Pan
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jiaojiao Yu
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China.
| | - Lehui Xiao
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China.
| | - Ran Cheng
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China.
| |
Collapse
|
6
|
Chauchat J, Turgay F, Kırmızıgil B, Yigittürk O, Durmaz M, Aşıkovalı S. Association between uncoupling protein 1-3826 A/G polymorphism and the effects of anaerobic exercises on serum oxidative stress. Sci Sports 2024; 39:472-482. [DOI: 10.1016/j.scispo.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
7
|
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
|
8
|
Meng Z, Liu C, Xu M, Tao Y, Li H, Wang X, Liao J, Wang M. Adipose transplantation improves metabolism and atherosclerosis but not perivascular adipose tissue abnormality or vascular dysfunction in lipodystrophic Seipin/Apoe null mice. Am J Physiol Cell Physiol 2024; 326:C1410-C1422. [PMID: 38525541 PMCID: PMC11371364 DOI: 10.1152/ajpcell.00698.2023] [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: 12/17/2023] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Adipose dysfunction in lipodystrophic SEIPIN deficiency is associated with multiple metabolic disorders and increased risks of developing cardiovascular diseases, such as atherosclerosis, cardiac hypertrophy, and heart failure. Recently, adipose transplantation has been found to correct adipose dysfunction and metabolic disorders in lipodystrophic Seipin knockout mice; however, whether adipose transplantation could improve lipodystrophy-associated cardiovascular consequences is still unclear. Here, we aimed to explore the effects of adipose transplantation on lipodystrophy-associated metabolic cardiovascular diseases in Seipin knockout mice crossed into atherosclerosis-prone apolipoprotein E (Apoe) knockout background. At 2 months of age, lipodystrophic Seipin/Apoe double knockout mice and nonlipodystrophic Apoe knockout controls were subjected to adipose transplantation or sham operation. Seven months later, mice were euthanized. Our data showed that although adipose transplantation had no significant impact on endogenous adipose atrophy or gene expression, it remarkably increased plasma leptin but not adiponectin concentration in Seipin/Apoe double knockout mice. This led to significantly reduced hyperlipidemia, hepatic steatosis, and insulin resistance in Seipin/Apoe double knockout mice. Consequently, atherosclerosis burden, intraplaque macrophage infiltration, and aortic inflammatory gene expression were all attenuated in Seipin/Apoe double knockout mice with adipose transplantation. However, adipocyte morphology, macrophage infiltration, or fibrosis of the perivascular adipose tissue was not altered in Seipin/Apoe double knockout mice with adipose transplantation, followed by no significant improvement of vasoconstriction or relaxation. In conclusion, we demonstrate that adipose transplantation could alleviate lipodystrophy-associated metabolic disorders and atherosclerosis but has an almost null impact on perivascular adipose abnormality or vascular dysfunction in lipodystrophic Seipin/Apoe double knockout mice.NEW & NOTEWORTHY Adipose transplantation (AT) reverses multiply metabolic derangements in lipodystrophy, but whether it could improve lipodystrophy-related cardiovascular consequences is unknown. Here, using Seipin/Apoe double knockout mice as a lipodystrophy disease model, we showed that AT partially restored adipose functionality, which translated into significantly reduced atherosclerosis. However, AT was incapable of reversing perivascular adipose abnormality or vascular dysfunction. The current study provides preliminary experimental evidence on the therapeutic potential of AT on lipodystrophy-related metabolic cardiovascular diseases.
Collapse
Affiliation(s)
- Zhe Meng
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chuangxing Liu
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengke Xu
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yongqiang Tao
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyu Li
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xijia Wang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiawei Liao
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Mengyu Wang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Zheng Y, Li H, Bao Q, Tu Y, Ye Y, Jia W, Cao D. Brown Adipose Tissue Promotes Autologous Fat Grafts Retention Possibly Through Inhibiting Wnt/β-Catenin Pathway. Aesthetic Plast Surg 2024; 48:1817-1824. [PMID: 38409345 DOI: 10.1007/s00266-024-03888-4] [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: 08/25/2023] [Accepted: 01/30/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND In plastic surgery, autologous fat grafts (AFG) play an important role because of their abundant supply, biocompatibility, and low rejection rate. However, the lower retention rate of fat grafts limits their widespread use. Brown adipose tissue (BAT) can promote angiogenesis and regulate the level of associated inflammation. This study explored whether BAT has a facilitative effect on fat graft retention. METHODS We obtained white adipose tissue (WAT) from c57 mice and combined it with either BAT from c57 mice or phosphate-buffered saline (PBS) as a control. These mixtures were injected subcutaneously into the back of thymus-free nude mice. After 12 weeks, fat grafts were harvested, weighed, and analyzed. RESULTS We found that the BAT-grafted group had higher mass retention, more mature adipocytes, and higher vascularity than the other group. Further analysis revealed that BAT inhibited M1 macrophages; down-regulated IL-6, IL-1β, and TNF-β; upregulated M2 macrophages and Vascular endothelial growth factor-A (VEGFA); and promoted adipocyte regeneration by inhibiting the Wnt/β-catenin pathway, which together promoted adipose graft retention. CONCLUSION The study demonstrated that BAT improved adipose graft retention by promoting angiogenesis, inhibiting tissue inflammation levels and the Wnt/β-catenin pathway. LEVEL OF EVIDENCE III This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.
Collapse
Affiliation(s)
- Yunfeng Zheng
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital of Anhui Medical University, 678, FuRong Road, Hefei, 230601, Anhui, China
| | - Honghong Li
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital of Anhui Medical University, 678, FuRong Road, Hefei, 230601, Anhui, China
| | - Qiong Bao
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital of Anhui Medical University, 678, FuRong Road, Hefei, 230601, Anhui, China
| | - Yiqian Tu
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital of Anhui Medical University, 678, FuRong Road, Hefei, 230601, Anhui, China
| | - Yujie Ye
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital of Anhui Medical University, 678, FuRong Road, Hefei, 230601, Anhui, China
| | - Wenjun Jia
- Department of Breast Surgery, The Second Affiliated Hospital of Anhui Medical University, 678, FuRong Road, Hefei, 230601, Anhui, China.
| | - Dongsheng Cao
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital of Anhui Medical University, 678, FuRong Road, Hefei, 230601, Anhui, China.
| |
Collapse
|
10
|
Zhang Y, Xue J, Zhu W, Wang H, Xi P, Tian D. TRPV4 in adipose tissue ameliorates diet-induced obesity by promoting white adipocyte browning. Transl Res 2024; 266:16-31. [PMID: 37926276 DOI: 10.1016/j.trsl.2023.11.001] [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: 08/15/2023] [Revised: 10/12/2023] [Accepted: 11/02/2023] [Indexed: 11/07/2023]
Abstract
The induction of adipocyte browning to increase energy expenditure is a promising strategy to combat obesity. Transient receptor potential channel V4 (TRPV4) functions as a nonselective cation channel in various cells and plays physiological roles in osmotic and thermal sensations. However, the function of TRPV4 in energy metabolism remains controversial. This study revealed the role of TRPV4 in adipose tissue in the development of obesity. Adipose-specific TRPV4 overexpression protected mice against diet-induced obesity (DIO) and promoted white fat browning. TRPV4 overexpression was also associated with decreased adipose inflammation and improved insulin sensitivity. Mechanistically, TRPV4 could directly promote white adipocyte browning via the AKT pathway. Consistently, adipose-specific TRPV4 knockout exacerbated DIO with impaired thermogenesis and activated inflammation. Corroborating our findings in mice, TRPV4 expression was low in the white adipose tissue of obese people. Our results positioned TRPV4 as a potential regulator of obesity and energy expenditure in mice and humans.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China
| | - Jie Xue
- Department of Pathology, Handan Central Hospital, Handan, Hebei 057150, China
| | - Wenjuan Zhu
- Department of Nuclear Medicine, Third Hospital of Nanchang, Nanchang, Jiangxi 330008, China
| | - Haomin Wang
- Department of Human Anatomy and Histology, Tianjin Medical University, Tianjin 300070, China
| | - Pengjiao Xi
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China.
| | - Derun Tian
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China; Department of Human Anatomy and Histology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
11
|
Dewal RS, Yang FT, Baer LA, Vidal P, Hernandez-Saavedra D, Seculov NP, Ghosh A, Noé F, Togliatti O, Hughes L, DeBari MK, West MD, Soroko R, Sternberg H, Malik NN, Puchulu-Campanella E, Wang H, Yan P, Wolfrum C, Abbott RD, Stanford KI. Transplantation of committed pre-adipocytes from brown adipose tissue improves whole-body glucose homeostasis. iScience 2024; 27:108927. [PMID: 38327776 PMCID: PMC10847743 DOI: 10.1016/j.isci.2024.108927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/15/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024] Open
Abstract
Obesity and its co-morbidities including type 2 diabetes are increasing at epidemic rates in the U.S. and worldwide. Brown adipose tissue (BAT) is a potential therapeutic to combat obesity and type 2 diabetes. Increasing BAT mass by transplantation improves metabolic health in rodents, but its clinical translation remains a challenge. Here, we investigated if transplantation of 2-4 million differentiated brown pre-adipocytes from mouse BAT stromal fraction (SVF) or human pluripotent stem cells (hPSCs) could improve metabolic health. Transplantation of differentiated brown pre-adipocytes, termed "committed pre-adipocytes" from BAT SVF from mice or derived from hPSCs improves glucose homeostasis and insulin sensitivity in recipient mice under conditions of diet-induced obesity, and this improvement is mediated through the collaborative actions of the liver transcriptome, tissue AKT signaling, and FGF21. These data demonstrate that transplantation of a small number of brown adipocytes has significant long-term translational and therapeutic potential to improve glucose metabolism.
Collapse
Affiliation(s)
- Revati S. Dewal
- Department of Physiology and Cell Biology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Felix T. Yang
- Department of Physiology and Cell Biology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- Department of Surgery, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Lisa A. Baer
- Department of Physiology and Cell Biology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- Department of Surgery, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Pablo Vidal
- Department of Physiology and Cell Biology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- Department of Surgery, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Diego Hernandez-Saavedra
- Department of Physiology and Cell Biology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Nickolai P. Seculov
- Department of Physiology and Cell Biology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Adhideb Ghosh
- Laboratory of Translational Nutritional Biology, Institute of Food, Nutrition and Health, ETH Zurich, 8603 Schwerzenbach, Switzerland
| | - Falko Noé
- Laboratory of Translational Nutritional Biology, Institute of Food, Nutrition and Health, ETH Zurich, 8603 Schwerzenbach, Switzerland
| | - Olivia Togliatti
- Department of Physiology and Cell Biology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Lexis Hughes
- Department of Physiology and Cell Biology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Megan K. DeBari
- Department of Biomedical Engineering, College of Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Michael D. West
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Suite 201, Alameda, CA 94501, USA
| | - Richard Soroko
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Suite 201, Alameda, CA 94501, USA
| | - Hal Sternberg
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Suite 201, Alameda, CA 94501, USA
| | - Nafees N. Malik
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Suite 201, Alameda, CA 94501, USA
| | - Estella Puchulu-Campanella
- Genomics Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Huabao Wang
- Genomics Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Pearlly Yan
- Genomics Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Christian Wolfrum
- Laboratory of Translational Nutritional Biology, Institute of Food, Nutrition and Health, ETH Zurich, 8603 Schwerzenbach, Switzerland
| | - Rosalyn D. Abbott
- Department of Biomedical Engineering, College of Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Kristin I. Stanford
- Department of Physiology and Cell Biology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- Department of Surgery, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
12
|
Wang S, Xiao Y, An X, Luo L, Gong K, Yu D. A comprehensive review of the literature on CD10: its function, clinical application, and prospects. Front Pharmacol 2024; 15:1336310. [PMID: 38389922 PMCID: PMC10881666 DOI: 10.3389/fphar.2024.1336310] [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: 11/14/2023] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
CD10, a zinc-dependent metalloprotease found on the cell surface, plays a pivotal role in an array of physiological and pathological processes including cardiovascular regulation, immune function, fetal development, pain response, oncogenesis, and aging. Recognized as a biomarker for hematopoietic and tissue stem cells, CD10 has garnered attention for its prognostic potential in the progression of leukemia and various solid tumors. Recent studies underscore its regulatory significance and therapeutic promise in combating Alzheimer's disease (AD), and it is noted for its protective role in preventing heart failure (HF), obesity, and type-2 diabetes. Furthermore, CD10/substance P interaction has also been shown to contribute to the pain signaling regulation and immunomodulation in diseases such as complex regional pain syndrome (CRPS) and osteoarthritis (OA). The emergence of COVID-19 has sparked interest in CD10's involvement in the disease's pathogenesis. Given its association with multiple disease states, CD10 is a prime therapeutic target; inhibitors targeting CD10 are now being advanced as therapeutic agents. This review compiles recent and earlier literature on CD10, elucidating its physicochemical attributes, tissue-specific expression, and molecular functions. Furthermore, it details the association of CD10 with various diseases and the clinical advancements of its inhibitors, providing a comprehensive overview of its growing significance in medical research.
Collapse
Affiliation(s)
- Shudong Wang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yinghui Xiao
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xingna An
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ling Luo
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Kejian Gong
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dehai Yu
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
13
|
Kudo M, Gao M, Hayashi M, Kobayashi Y, Yang J, Liu T. Ilex paraguariensis A.St.-Hil. improves lipid metabolism in high-fat diet-fed obese rats and suppresses intracellular lipid accumulation in 3T3-L1 adipocytes via the AMPK-dependent and insulin signaling pathways. Food Nutr Res 2024; 68:10307. [PMID: 38327997 PMCID: PMC10845893 DOI: 10.29219/fnr.v68.10307] [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: 10/19/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 02/09/2024] Open
Abstract
Background Obesity is closely associated with several chronic diseases, and adipose tissue plays a major role in modulating energy metabolism. Objective This study aimed to determine whether Mate, derived from I. paraguariensis A.St.-Hil., ameliorates lipid metabolism in 3T3-L1 adipocytes and high-fat diet (HFD)-fed obese Sprague-Dawley (SD) rats. Design 3T3-L1 adipocytes were cultured for 7 days, following which intracellular lipid accumulation and expression levels of lipid metabolism-related factors were examined. Dorsomorphin was used to investigate the potential pathways involved, particularly the adenosine monophosphate-activated protein kinase (AMPK)- dependent pathway. Mate was administered to rat HFD-fed obese SD models for 8 consecutive weeks. The expression of lipid metabolism-related factors in the organs and tissues collected from dissected SD rats was evaluated. Results Mate suppressed intracellular lipid accumulation in 3T3-L1 adipocytes, increased the protein and gene expression levels of AMPK, hormone sensitive lipase (HSL), calmodulin kinase kinase (CaMKK), liver kinase B1 (LKB1), protein kinase A (PKA), CCAAT/enhancer binding protein β (C/EBPβ), insulin receptor b (IRβ), and insulin receptor substrate 1 (IRS1) (Tyr465), and decreased those of sterol regulatory element binding protein 1C (Srebp1c), fatty acid synthase (FAS), peroxisome-activated receptor γ (PPARγ), and IRS1 (Ser1101). Furthermore, an AMPK inhibitor abolished the effects exerted by Mate on intracellular lipid accumulation and HSL and FAS expression levels. Mate treatment suppressed body weight gain and improved serum cholesterol levels in HFD-fed obese SD rats. Treatment with Mate increased the protein and gene expression levels of AMPK, PKA, Erk1/Erk2 (p44/p42), and uncoupling protein 1 and reduced those of mammalian target of rapamycin, S6 kinase, Srebp1c, ap2, FAS, Il6, Adiponectin, Leptin, and Fabp4 in rat HFD-fed obese SD models. Discussion and conclusions Mate suppressed intracellular lipid accumulation in 3T3-L1 adipocytes and improved lipid metabolism in the epididymal adipose tissue of HFD-fed obese SD rats via the activation of AMPK-dependent and insulin signaling pathways.
Collapse
Affiliation(s)
- Maya Kudo
- School of Pharmacy and Pharmaceutical Science, Mukogawa Women’s University, Nishinomiya, Hyogo, Japan
| | - Ming Gao
- School of Pharmacy and Pharmaceutical Science, Mukogawa Women’s University, Nishinomiya, Hyogo, Japan
- Institute for Bioscience, Mukogawa Women’s University, Nishinomiya, Hyogo, Japan
| | - Misa Hayashi
- School of Pharmacy and Pharmaceutical Science, Mukogawa Women’s University, Nishinomiya, Hyogo, Japan
| | | | - Jinwei Yang
- Tokiwa Phytochemical Co., Ltd., Sakura, Chiba, Japan
| | - Tonghua Liu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Mahmoud M, Abdel-Rasheed M, Galal ER, El-Awady RR. Factors Defining Human Adipose Stem/Stromal Cell Immunomodulation in Vitro. Stem Cell Rev Rep 2024; 20:175-205. [PMID: 37962697 PMCID: PMC10799834 DOI: 10.1007/s12015-023-10654-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 11/15/2023]
Abstract
Human adipose tissue-derived stem/stromal cells (hASCs) are adult multipotent mesenchymal stem/stromal cells with immunomodulatory capacities. Here, we present up-to-date knowledge on the impact of different experimental and donor-related factors on hASC immunoregulatory functions in vitro. The experimental determinants include the immunological status of hASCs relative to target immune cells, contact vs. contactless interaction, and oxygen tension. Factors such as the ratio of hASCs to immune cells, the cellular context, the immune cell activation status, and coculture duration are also discussed. Conditioning of hASCs with different approaches before interaction with immune cells, hASC culture in xenogenic or xenofree culture medium, hASC culture in two-dimension vs. three-dimension with biomaterials, and the hASC passage number are among the experimental parameters that greatly may impact the hASC immunosuppressive potential in vitro, thus, they are also considered. Moreover, the influence of donor-related characteristics such as age, sex, and health status on hASC immunomodulation in vitro is reviewed. By analysis of the literature studies, most of the indicated determinants have been investigated in broad non-standardized ranges, so the results are not univocal. Clear conclusions cannot be drawn for the fine-tuned scenarios of many important factors to set a standard hASC immunopotency assay. Such variability needs to be carefully considered in further standardized research. Importantly, field experts' opinions may help to make it clearer.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt.
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt.
| | - Mazen Abdel-Rasheed
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt
- Department of Reproductive Health Research, National Research Centre, Cairo, Egypt
| | - Eman Reda Galal
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Rehab R El-Awady
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
15
|
Rui C, Chan MKS, Skutella T. Stem Cell Therapies and Ageing: Unlocking the Potential of Regenerative Medicine. Subcell Biochem 2024; 107:117-128. [PMID: 39693022 DOI: 10.1007/978-3-031-66768-8_6] [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] [Indexed: 12/19/2024]
Abstract
A multifaceted biological process of ageing culminates in the gradual decline of tissue and organ functions, escalating vulnerability to age-related diseases. Stem cell therapies, standing at the frontier of regenerative medicine, hold the potential to mitigate the challenges induced by ageing. By harnessing the unique regenerative capabilities of stem cells, these therapies aim to renew and heal ageing or damaged cells and tissues, thereby bolstering their function. In this chapter, we explore the potential of stem cell-based interventions against age-related degeneration, emphasising their underlying mechanisms, challenges, and future possibilities. As elucidated by the Buck Institute for Research on Aging, ageing is characterised by an accrual of macromolecular damage, genomic instability, and loss of heterochromatin (Campisi et al. Nature 571:183-192, 2019). These aspects culminate in stem cell fatigue and a dwindling tissue regenerative capacity. However, with the advent of stem cell therapy and regenerative medicine, we now hold the tools to reverse these age-induced changes by rejuvenating stem cells, the keystones of tissue regeneration, and fostering their proliferation and differentiation.
Collapse
Affiliation(s)
- Chen Rui
- Reproductive Medicine Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Thomas Skutella
- Group for Regeneration and Reprogramming, Institute for Anatomy and Cell Biology, Medical Faculty, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
16
|
Rabadán-Chávez G, Díaz de la Garza RI, Jacobo-Velázquez DA. White adipose tissue: Distribution, molecular insights of impaired expandability, and its implication in fatty liver disease. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166853. [PMID: 37611674 DOI: 10.1016/j.bbadis.2023.166853] [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: 04/15/2023] [Revised: 07/17/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
We are far behind the 2025 World Health Organization (WHO) goal of a zero increase in obesity. Close to 360 million people in Latin America and the Caribbean are overweight, with the highest rates observed in the Bahamas, Mexico, and Chile. To achieve relevant progress against the obesity epidemic, scientific research is essential to establish uniform practices in the study of obesity pathophysiology (using pre-clinical and clinical models) that ensure accuracy, reproducibility, and transcendent outcomes. The present review focuses on relevant aspects of white adipose tissue (WAT) expansion, underlying mechanisms of inefficient expandability, and its repercussion in ectopic lipid accumulation in the liver during nutritional abundance. In addition, we highlight the potential role of disrupted circadian rhythm in WAT metabolism. Since genetic factors also play a key role in determining an individual's predisposition to weight gain, we describe the most relevant genes associated with obesity in the Mexican population, underlining that most of them are related to appetite control.
Collapse
Affiliation(s)
- Griselda Rabadán-Chávez
- Tecnologico de Monterrey, Institute for Obesity Research, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico
| | - Rocío I Díaz de la Garza
- Tecnologico de Monterrey, Institute for Obesity Research, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico; Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Monterrey, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico.
| | - Daniel A Jacobo-Velázquez
- Tecnologico de Monterrey, Institute for Obesity Research, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico; Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Guadalajara, Av. General Ramon Corona 2514, C.P. 45201 Zapopan, Jalisco, Mexico.
| |
Collapse
|
17
|
Chai J, Wang N, Chen L, Bai J, Zhang J, Zhang G, An J, Zhang T, Tong X, Wu Y, Li M, Jin L. Identification of a Novel Long Non-Coding RNA G8110 That Modulates Porcine Adipogenic Differentiation and Inflammatory Responses. Int J Mol Sci 2023; 24:16799. [PMID: 38069122 PMCID: PMC10706401 DOI: 10.3390/ijms242316799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/14/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have been extensively studied, and their crucial roles in adipogenesis, lipid metabolism, and gene expression have been revealed. However, the exact regulatory or other mechanisms by which lncRNAs influence the functioning of mesenteric adipose tissue (MAT) remain largely unknown. In this paper, we report the identification of a new lncRNA, named G8110, from the MAT of Bama pigs. The coordinated expression levels of lncRNA G8110 and NFE2L1 were significantly decreased in the MAT of obese Bama pigs compared with those in the MAT of lean pigs. Using a bone mesenchymal stem cell adipogenic differentiation model, we found that lncRNA G8110 played a role in adipocyte differentiation by positively regulating NFE2L1. We also found that lncRNA G8110 inhibited the formation of intracellular lipid synthesis, promoted lipid metabolism, and inhibited the expression of inflammatory cytokines. Our findings regarding lipid synthesis may further promote the role of lncRNAs in driving adipose tissue remodeling and maintaining metabolic health.
Collapse
Affiliation(s)
- Jin Chai
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.C.); (N.W.); (J.B.); (J.Z.); (G.Z.); (J.A.); (T.Z.); (X.T.); (Y.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Ning Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.C.); (N.W.); (J.B.); (J.Z.); (G.Z.); (J.A.); (T.Z.); (X.T.); (Y.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Chen
- Chongqing Academy of Animal Science, Chongqing 402460, China;
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
- Key Laboratory of Animal Resource Evaluation and Utilization (Pigs), Ministry of Agriculture and Rural Affairs, Chongqing 402160, China
| | - Jingyi Bai
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.C.); (N.W.); (J.B.); (J.Z.); (G.Z.); (J.A.); (T.Z.); (X.T.); (Y.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiaman Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.C.); (N.W.); (J.B.); (J.Z.); (G.Z.); (J.A.); (T.Z.); (X.T.); (Y.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Geng Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.C.); (N.W.); (J.B.); (J.Z.); (G.Z.); (J.A.); (T.Z.); (X.T.); (Y.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiahua An
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.C.); (N.W.); (J.B.); (J.Z.); (G.Z.); (J.A.); (T.Z.); (X.T.); (Y.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Tingting Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.C.); (N.W.); (J.B.); (J.Z.); (G.Z.); (J.A.); (T.Z.); (X.T.); (Y.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xingyan Tong
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.C.); (N.W.); (J.B.); (J.Z.); (G.Z.); (J.A.); (T.Z.); (X.T.); (Y.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yifan Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.C.); (N.W.); (J.B.); (J.Z.); (G.Z.); (J.A.); (T.Z.); (X.T.); (Y.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingzhou Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.C.); (N.W.); (J.B.); (J.Z.); (G.Z.); (J.A.); (T.Z.); (X.T.); (Y.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Long Jin
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.C.); (N.W.); (J.B.); (J.Z.); (G.Z.); (J.A.); (T.Z.); (X.T.); (Y.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
18
|
Wang H, Yu L, Wang J, Zhang Y, Xu M, Lv C, Cui B, Yuan M, Zhang Y, Yan Y, Hui R, Wang Y. SLC35D3 promotes white adipose tissue browning to ameliorate obesity by NOTCH signaling. Nat Commun 2023; 14:7643. [PMID: 37996411 PMCID: PMC10667520 DOI: 10.1038/s41467-023-43418-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
White adipose tissue browning can promote lipid burning to increase energy expenditure and improve adiposity. Here, we show that Slc35d3 expression is significantly lower in adipose tissues of obese mice. While adipocyte-specific Slc35d3 knockin is protected against diet-induced obesity, adipocyte-specific Slc35d3 knockout inhibits white adipose tissue browning and causes decreased energy expenditure and impaired insulin sensitivity in mice. Mechanistically, we confirm that SLC35D3 interacts with the NOTCH1 extracellular domain, which leads to the accumulation of NOTCH1 in the endoplasmic reticulum and thus inhibits the NOTCH1 signaling pathway. In addition, knockdown of Notch1 in mouse inguinal white adipose tissue mediated by orthotopic injection of AAV8-adiponectin-shNotch1 shows considerable improvement in obesity and glucolipid metabolism, which is more pronounced in adipocyte-specific Slc35d3 knockout mice than in knockin mice. Overall, in this study, we reveal that SLC35D3 is involved in obesity via NOTCH1 signaling, and low adipose SLC35D3 expression in obesity might be a therapeutic target for obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Hongrui Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jin'e Wang
- College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Yaqing Zhang
- College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Mengchen Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng Lv
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing Cui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengmeng Yuan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yupeng Yan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rutai Hui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yibo Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
19
|
Li Y, Zhang Y, Zhang T, Ping X, Wang D, Chen Y, Yu J, Liu C, Liu Z, Zheng Y, Yang Y, Ruan C, Li D, Du Z, Wang J, Xu L, Ma X. Rna M 6 a Methylation Regulates Glycolysis of Beige Fat and Contributes to Systemic Metabolic Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300436. [PMID: 37407508 PMCID: PMC10477848 DOI: 10.1002/advs.202300436] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/29/2023] [Indexed: 07/07/2023]
Abstract
N6-methyladenosine (m6 A) modification has been implicated in the progression of obesity and metabolic diseases. However, its impact on beige fat biology is not well understood. Here, via m6 A-sequencing and RNA-sequencing, this work reports that upon beige adipocytes activation, glycolytic genes undergo major events of m6 A modification and transcriptional activation. Genetic ablation of m6 A writer Mettl3 in fat tissues reveals that Mettl3 deficiency in mature beige adipocytes leads to suppressed glycolytic capability and thermogenesis, as well as reduced preadipocytes proliferation via glycolytic product lactate. In addition, specific modulation of Mettl3 in beige fat via AAV delivery demonstrates consistently Mettl3's role in glucose metabolism, thermogenesis, and beige fat hyperplasia. Mechanistically, Mettl3 and m6 A reader Igf2bp2 control mRNA stability of key glycolytic genes in beige adipocytes. Overall, these findings highlight the significance of m6 A on fat biology and systemic energy homeostasis.
Collapse
Affiliation(s)
- Yu Li
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
- Chongqing Key Laboratory of Precision OpticsChongqing Institute of East China Normal UniversityChongqing401120China
| | - Yankang Zhang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Ting Zhang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Xiaodan Ping
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Dongmei Wang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Yanru Chen
- Department of Endocrinology and MetabolismRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Jian Yu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
- Department of Endocrinology and MetabolismFengxian Central Hospital Affiliated to Southern Medical UniversityShanghai201499China
| | - Caizhi Liu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Ziqi Liu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Yuhan Zheng
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Yongfeng Yang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Chengchao Ruan
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory Biology and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Zhenyu Du
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Jiqiu Wang
- Department of Endocrinology and MetabolismRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
- Chongqing Key Laboratory of Precision OpticsChongqing Institute of East China Normal UniversityChongqing401120China
- Department of Endocrinology and MetabolismFengxian Central Hospital Affiliated to Southern Medical UniversityShanghai201499China
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory Biology and School of Life SciencesEast China Normal UniversityShanghai200241China
| |
Collapse
|
20
|
Huang Y, Cui D, Chen L, Tong H, Wu H, Muller GK, Qi Y, Wang S, Xu J, Gao X, Fifield KE, Wang L, Xia Z, Vanderluit JL, Liu S, Leng L, Sun G, McGuire J, Young LH, Bucala R, Qi D. A pref-1-controlled non-inflammatory mechanism of insulin resistance. iScience 2023; 26:106923. [PMID: 37283810 PMCID: PMC10239698 DOI: 10.1016/j.isci.2023.106923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/21/2023] [Accepted: 05/15/2023] [Indexed: 06/08/2023] Open
Abstract
While insulin resistance (IR) is associated with inflammation in white adipose tissue, we report a non-inflammatory adipose mechanism of high fat-induced IR mediated by loss of Pref-1. Pref-1, released from adipose Pref-1+ cells with characteristics of M2 macrophages, endothelial cells or progenitors, inhibits MIF release from both Pref-1+ cells and adipocytes by binding with integrin β1 and inhibiting the mobilization of p115. High palmitic acid induces PAR2 expression in Pref-1+ cells, downregulating Pref-1 expression and release in an AMPK-dependent manner. The loss of Pref-1 increases adipose MIF secretion contributing to non-inflammatory IR in obesity. Treatment with Pref-1 blunts the increase in circulating plasma MIF levels and subsequent IR induced by a high palmitic acid diet. Thus, high levels of fatty acids suppress Pref-1 expression and secretion, through increased activation of PAR2, resulting in an increase in MIF secretion and a non-inflammatory adipose mechanism of IR.
Collapse
Affiliation(s)
- Yiheng Huang
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Donghong Cui
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liujun Chen
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Haibin Tong
- College of Life and Environment Sciences, Wenzhou University, Wenzhou, Zhejiang, China
| | - Hong Wu
- Institute of Cardiovascular Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Grace K. Muller
- Department of Cell and Molecular Physiology, Loyola University, Chicago, IL, USA
| | - Yadan Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Shuxia Wang
- Department of Cardiology, The General Hospital of Chinese PLA, Beijing, China
| | - Jinjie Xu
- Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Xiang Gao
- College of Life Sciences, Qingdao University, Qingdao, Shandong, China
| | - Kathleen E. Fifield
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Lingyan Wang
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | | | - Jacqueline L. Vanderluit
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Suixin Liu
- Division of Cardiac Rehabilitation, Department of Physical Medicine & Rehabilitation, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Guang Sun
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - John McGuire
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Lawrence H. Young
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Dake Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| |
Collapse
|
21
|
Zhang H, Mi S, Brito LF, Hu L, Wang L, Ma L, Xu Q, Guo G, Yu Y, Wang Y. Genomic and transcriptomic analyses enable the identification of important genes associated with subcutaneous fat deposition in Holstein cows. J Genet Genomics 2023; 50:385-397. [PMID: 36738887 DOI: 10.1016/j.jgg.2023.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/05/2023]
Abstract
Subcutaneous fat deposition has many important roles in dairy cattle, including immunological defense and mechanical protection. The main objectives of this study are to identify key candidate genes regulating subcutaneous fat deposition in high-producing dairy cows by integrating genomic and transcriptomic datasets. A total of 1654 genotyped Holstein cows are used to perform a genome-wide association study (GWAS) aiming to identify genes associated with subcutaneous fat deposition. Subsequently, weighted gene co-expression network analyses (WGCNA) are conducted based on RNA-sequencing data of 34 cows and cow yield deviations of subcutaneous fat deposition. Lastly, differentially expressed (DE) mRNA, lncRNA, and differentially alternative splicing genes are obtained for 12 Holstein cows with extreme and divergent phenotypes for subcutaneous fat deposition. Forty-six protein-coding genes are identified as candidate genes regulating subcutaneous fat deposition in Holstein cattle based on GWAS. Eleven overlapping genes are identified based on the analyses of DE genes and WGCNA. Furthermore, the candidate genes identified based on GWAS, WGCNA, and analyses of DE genes are significantly enriched for pathways involved in metabolism, oxidative phosphorylation, thermogenesis, fatty acid degradation, and glycolysis/gluconeogenesis pathways. Integrating all findings, the NID2, STARD3, UFC1, DEDD, PPP1R1B, and USP21 genes are considered to be the most important candidate genes influencing subcutaneous fat deposition traits in Holstein cows. This study provides novel insights into the regulation mechanism underlying fat deposition in high-producing dairy cows, which will be useful when designing management and breeding strategies.
Collapse
Affiliation(s)
- Hailiang Zhang
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Siyuan Mi
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Lirong Hu
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Lei Wang
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Longgang Ma
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Qing Xu
- Institute of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| | - Gang Guo
- Beijing Sunlon Livestock Development Co. Ltd, Beijing 100176, China
| | - Ying Yu
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yachun Wang
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
22
|
Parascandolo A, Di Tolla MF, Liguoro D, Lecce M, Misso S, Micieli F, Ambrosio MR, Cabaro S, Beguinot F, Pelagalli A, D'Esposito V, Formisano P. Human Platelet-Rich Plasma Regulates Canine Mesenchymal Stem Cell Migration through Aquaporins. Stem Cells Int 2023; 2023:8344259. [PMID: 37223543 PMCID: PMC10202607 DOI: 10.1155/2023/8344259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 04/19/2023] [Accepted: 05/05/2023] [Indexed: 05/25/2023] Open
Abstract
Platelet products are commonly used in regenerative medicine due to their effects on the acceleration and promotion of wound healing, reduction of bleeding, synthesis of new connective tissue, and revascularization. Furthermore, a novel approach for the treatment of damaged tissues, following trauma or other pathological damages, is represented by the use of mesenchymal stem cells (MSCs). In dogs, both platelet-rich plasma (PRP) and MSCs have been suggested to be promising options for subacute skin wounds. However, the collection of canine PRP is not always feasible. In this study, we investigated the effect of human PRP (hPRP) on canine MSCs (cMSCs). We isolated cMSCs and observed that hPRP did not modify the expression levels of the primary class of major histocompatibility complex genes. However, hPRP was able to increase cMSC viability and migration by at least 1.5-fold. hPRP treatment enhanced both Aquaporin (AQP) 1 and AQP5 protein levels, and their inhibition by tetraethylammonium chloride led to a reduction of PRP-induced migration of cMSCs. In conclusion, we have provided evidence that hPRP supports cMSC survival and may promote cell migration, at least through AQP activation. Thus, hPRP may be useful in canine tissue regeneration and repair, placing as a promising tool for veterinary therapeutic approaches.
Collapse
Affiliation(s)
- Alessia Parascandolo
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Michele Francesco Di Tolla
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Domenico Liguoro
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Manuela Lecce
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Saverio Misso
- Unit of Transfusion Medicine, Azienda Sanitaria Locale Caserta, Caserta, Italy
| | - Fabiana Micieli
- Department of Veterinary Medicine and Animal Productions, University of Napoli Federico II, 80137 Naples, Italy
| | - Maria Rosaria Ambrosio
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Serena Cabaro
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Napoli Federico II, 80131 Naples, Italy
- Institute of Biostructures and Bioimages, National Research Council, 80145 Naples, Italy
| | - Vittoria D'Esposito
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
23
|
Ambrosio MR, Migliaccio T, Napolitano F, Di Somma S, Maneli G, Amato J, Pagano B, Randazzo A, Portella G, Formisano P, Malfitano AM. Targeting G-quadruplex motifs interferes with differentiation of adipose-derived mesenchymal stem cells. Stem Cell Res Ther 2023; 14:98. [PMID: 37076894 PMCID: PMC10116735 DOI: 10.1186/s13287-023-03320-9] [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: 11/24/2022] [Accepted: 03/29/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND G-quadruplex (G4) motifs are nucleic acid secondary structures observed in mammalian genomes and transcriptomes able to regulate various cellular processes. Several small molecules have been developed so far to modulate G4 stability, frequently associated with anticancer activity. However, how G4 structures are regulated over homeostatic conditions is mostly unexplored. Here, we used human adipose-derived mesenchymal stem cells (ASCs) to address the role of G4 motifs during adipogenic differentiation. METHODS Adipocyte differentiation of ASCs was investigated in the presence or absence of a well-known G4 ligand, Braco-19. Cell viability was determined by sulforhodamine B assay. Cell dimension and granularity, DNA G4 motifs and cell cycle were detected by flow cytometry. Lipid droplet accumulation was assessed by Oil Red O staining. Cell senescence was evaluated by β-galactosidase staining. Gene expression was measured by qPCR. Protein release in the extracellular medium was quantified by ELISA. RESULTS Braco-19 used at non-cytotoxic concentrations induced morphological changes in mature adipocytes partially restoring an undifferentiated-like status. Braco-19 reduced lipid vacuolization and PPARG, AP2, LEP and TNFA mRNA levels in terminally differentiated cells. No effect was observed in cell senescence, fibrotic markers, IL-6 and IL-8 production, while the secretion of VEGF was dose-dependently reduced. Interestingly, G4 structures were increased in differentiated adipocytes compared to their precursors. Braco-19 treatment reduced G4 content in mature adipocytes. CONCLUSIONS Our data highlight a new role of G4 motifs as genomic structural elements related to human ASC differentiation into mature adipocytes, with potential implications in physio-pathological processes.
Collapse
Affiliation(s)
- Maria Rosaria Ambrosio
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - Teresa Migliaccio
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Fabiana Napolitano
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Sarah Di Somma
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Giovanni Maneli
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Jussara Amato
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Bruno Pagano
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Antonio Randazzo
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Giuseppe Portella
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy.
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy.
| | - Anna Maria Malfitano
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy.
| |
Collapse
|
24
|
Tan M, Xu H, Li J, Jia Z, Zhang X, Shao S, Zhang W, Wang W, Sun Y. PU.1 interacts with KLF7 to suppress differentiation and promote proliferation in chicken preadipocytes. Acta Biochim Biophys Sin (Shanghai) 2023; 55:143-153. [PMID: 36647727 PMCID: PMC10157628 DOI: 10.3724/abbs.2022202] [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: 01/01/2023] Open
Abstract
<p indent="0mm">Krüppel-like factor 7 (KLF7) is a negative regulator of preadipocyte differentiation. Our previous KLF7 ChIP-seq analysis showed that the binding motif of PU.1 was found among the KLF7 binding peaks, indicating that an interaction between KLF7 and PU.1 at preadipocyte gene promoters and other regulatory elements might be common. Here, Co-IP and FRET assays are used to confirm that PU.1 can directly bind to KLF7 and enhance the transcription activity of cyclin-dependent kinase inhibitor 3 ( <italic>CDKN3</italic>), which is a downstream target gene of KLF7. We show that the PU.1 expression level is decreased during preadipocyte differentiation. Furthermore, PU.1 overexpression and knockdown experiments reveal that PU.1 negatively regulates chicken preadipocyte differentiation, as evidenced by appropriate changes in lipid droplet accumulation and altered expressions of PPARγ, FAS, and PLIN. In addition, PU.1 overexpression promotes preadipocyte proliferation, while knockdown of <italic>PU</italic>. <italic>1</italic> inhibits preadipocyte proliferation. We further demonstrate that PU.1 inhibits differentiation and promotes proliferation in preadipocytes, in part by directly interacting with KLF7. </p>.
Collapse
|
25
|
Biomimetic nanofiber-enabled rapid creation of skin grafts. Nanomedicine (Lond) 2023. [DOI: 10.1016/b978-0-12-818627-5.00009-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
|
26
|
Affiliation(s)
- Dan Xu
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
27
|
Daquinag AC, Gao Z, Yu Y, Kolonin MG. Endothelial TrkA coordinates vascularization and innervation in thermogenic adipose tissue and can be targeted to control metabolism. Mol Metab 2022; 63:101544. [PMID: 35835372 PMCID: PMC9310128 DOI: 10.1016/j.molmet.2022.101544] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/24/2022] [Accepted: 07/05/2022] [Indexed: 10/25/2022] Open
Abstract
OBJECTIVE Brown adipogenesis and thermogenesis in brown and beige adipose tissue (AT) involve vascular remodeling and sympathetic neuronal guidance. Here, we investigated the molecular mechanism coordinating these processes. METHODS We used mouse models to identify the molecular target of a peptide CPATAERPC homing to the endothelium of brown and beige AT. RESULTS We demonstrate that CPATAERPC mimics nerve growth factor (NGF) and identify a low molecular weight isoform of NGF receptor, TrkA, as the CPATAERPC cell surface target. We show that the expression of truncated endothelial TrkA is selective for brown and subcutaneous AT. Analysis of mice with endothelium-specific TrkA knockout revealed the role of TrkA in neuro-vascular coordination supporting the thermogenic function of brown adipocytes. A hunter-killer peptide D-BAT, composed of CPATAERPC and a pro-apoptotic domain, induced cell death in the endothelium and adipocytes. This resulted in thermogenesis impairment, and predisposed mice to obesity and glucose intolerance. We also tested if this treatment can inhibit the tumor recruitment of lipids mobilized from adipocytes from adjacent AT. Indeed, in a mouse model of breast cancer D-BAT suppressed tumor-associated AT lipolysis, which resulted in reduced fatty acid utilization by cancer cells. CONCLUSION Our study demonstrates that TrkA signaling in the endothelium supports neuro-vascular coordination enabling beige adipogenesis.
Collapse
Affiliation(s)
- Alexes C Daquinag
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Zhanguo Gao
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA.
| |
Collapse
|
28
|
Wu M, Huang Y, Zhu Q, Zhu X, Xue L, Xiong J, Chen Y, Wu C, Guo Y, Li Y, Wu M, Wang S. Adipose tissue and ovarian aging: Potential mechanism and protective strategies. Ageing Res Rev 2022; 80:101683. [PMID: 35817297 DOI: 10.1016/j.arr.2022.101683] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/29/2022] [Accepted: 07/05/2022] [Indexed: 11/01/2022]
Abstract
Ovarian aging occurs approximately 10 years prior to the natural age-associated functional decline of other organ systems. With the increase of life expectancy worldwide, ovarian aging has gradually become a key health problem among women. Therefore, understanding the causes and molecular mechanisms of ovarian aging is very essential for the inhibition of age-related diseases and the promotion of health and longevity in women. Recently, studies have revealed an association between adipose tissue (AT) and ovarian aging. Alterations in the function and quantity of AT have profound consequences on ovarian function because AT is central for follicular development, lipid metabolism, and hormonal regulation. Moreover, the interplay between AT and the ovary is bidirectional, with ovary-derived signals directly affecting AT biology. In this review, we summarize the current knowledge of the complex molecular mechanisms controlling the crosstalk between the AT and ovarian aging, and further discuss how therapeutic targeting of the AT can delay ovarian aging.
Collapse
Affiliation(s)
- Meng Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Yibao Huang
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Qingqing Zhu
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Xiaoran Zhu
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Liru Xue
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ying Chen
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Chuqing Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Yican Guo
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Yinuo Li
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Mingfu Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
| | - Shixuan Wang
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
| |
Collapse
|
29
|
Katsouda A, Valakos D, Dionellis VS, Bibli SI, Akoumianakis I, Karaliota S, Zuhra K, Fleming I, Nagahara N, Havaki S, Gorgoulis VG, Thanos D, Antoniades C, Szabo C, Papapetropoulos A. MPST sulfurtransferase maintains mitochondrial protein import and cellular bioenergetics to attenuate obesity. J Exp Med 2022; 219:e20211894. [PMID: 35616614 PMCID: PMC9143789 DOI: 10.1084/jem.20211894] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/16/2022] [Accepted: 04/27/2022] [Indexed: 11/04/2022] Open
Abstract
Given the clinical, economic, and societal impact of obesity, unraveling the mechanisms of adipose tissue expansion remains of fundamental significance. We previously showed that white adipose tissue (WAT) levels of 3-mercaptopyruvate sulfurtransferase (MPST), a mitochondrial cysteine-catabolizing enzyme that yields pyruvate and sulfide species, are downregulated in obesity. Here, we report that Mpst deletion results in fat accumulation in mice fed a high-fat diet (HFD) through transcriptional and metabolic maladaptation. Mpst-deficient mice on HFD exhibit increased body weight and inguinal WAT mass, reduced metabolic rate, and impaired glucose/insulin tolerance. At the molecular level, Mpst ablation activates HIF1α, downregulates subunits of the translocase of outer/inner membrane (TIM/TOM) complex, and impairs mitochondrial protein import. MPST deficiency suppresses the TCA cycle, oxidative phosphorylation, and fatty acid oxidation, enhancing lipid accumulation. Sulfide donor administration to obese mice reverses the HFD-induced changes. These findings reveal the significance of MPST for white adipose tissue biology and metabolic health and identify a potential new therapeutic target for obesity.
Collapse
Affiliation(s)
- Antonia Katsouda
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Valakos
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Sofia-Iris Bibli
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
- German Centre for Cardiovascular Research Partner Site Rhein-Main, Frankfurt am Main, Germany
| | - Ioannis Akoumianakis
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Sevasti Karaliota
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute/National Institutes of Health, Frederick, MD
| | - Karim Zuhra
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
- German Centre for Cardiovascular Research Partner Site Rhein-Main, Frankfurt am Main, Germany
| | | | - Sophia Havaki
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassilis G. Gorgoulis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Thanos
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
30
|
Comparison of Biological Features of Wild European Rabbit Mesenchymal Stem Cells Derived from Different Tissues. Int J Mol Sci 2022; 23:ijms23126420. [PMID: 35742872 PMCID: PMC9224375 DOI: 10.3390/ijms23126420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 12/11/2022] Open
Abstract
Although the European rabbit is an "endangered" species and a notorious biological model, the analysis and comparative characterization of new tissue sources of rabbit mesenchymal stem cells (rMSCs) have not been well addressed. Here, we report for the first time the isolation and characterization of rMSCs derived from an animal belonging to a natural rabbit population within the native region of the species. New rMSC lines were isolated from different tissues: oral mucosa (rOM-MSC), dermal skin (rDS-MSC), subcutaneous adipose tissue (rSCA-MSC), ovarian adipose tissue (rOA-MSC), oviduct (rO-MSC), and mammary gland (rMG-MSC). The six rMSC lines showed plastic adhesion with fibroblast-like morphology and were all shown to be positive for CD44 and CD29 expression (characteristic markers of MSCs), and negative for CD34 or CD45 expression. In terms of pluripotency features, all rMSC lines expressed NANOG, OCT4, and SOX2. Furthermore, all rMSC lines cultured under osteogenic, chondrogenic, and adipogenic conditions showed differentiation capacity. In conclusion, this study describes the isolation and characterization of new rabbit cell lines from different tissue origins, with a clear mesenchymal pattern. We show that rMSC do not exhibit differences in terms of morphological features, expression of the cell surface, and intracellular markers of pluripotency and in vitro differentiation capacities, attributable to their tissue of origin.
Collapse
|
31
|
Dai W, Liu X, Su H, Li X, Xu Y, Yu Y. Influence of adipose tissue immune dysfunction on childhood obesity. Cytokine Growth Factor Rev 2022; 65:27-38. [PMID: 35595599 DOI: 10.1016/j.cytogfr.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022]
Abstract
In recent decades, a dramatic rise has been observed in the prevalence of obesity in childhood and adolescence, along with an increase in fetal microsomia rates. The increased risk of obesity during this key period in development negatively affects the health of the individual later in life. Immune cells residing and recruited to white adipose tissue have been highlighted as important factors contributing to the pathogenesis of childhood obesity. Immune dysfunction in the context of obesity begins early in childhood, which is different from the pathological characteristics and influencing factors of adipose immunity in adults. Here, we explore the current understanding of the roles of childhood and early life events that result in high risks for obesity by influencing adipose tissue immune dysfunction under the pathological condition of obesity. Such knowledge will help in determining the mechanisms of childhood and early life obesity in efforts to ameliorate chronic inflammation-related metabolic diseases.
Collapse
Affiliation(s)
- Wanlin Dai
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiyan Liu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Han Su
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Xuan Li
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Yingxi Xu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Yang Yu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
32
|
Sun W, Zhang T, Hu S, Tang Q, Long X, Yang X, Gun S, Chen L. Chromatin accessibility landscape of stromal subpopulations reveals distinct metabolic and inflammatory features of porcine subcutaneous and visceral adipose tissue. PeerJ 2022; 10:e13250. [PMID: 35646489 PMCID: PMC9138157 DOI: 10.7717/peerj.13250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/21/2022] [Indexed: 01/13/2023] Open
Abstract
Background Fat accumulation in visceral adipose tissue (VAT) confers increased risk for metabolic disorders of obesity, whereas accumulation of subcutaneous adipose tissue (SAT) is associated with lower risk and may be protective. Previous studies have shed light on the gene expression profile differences between SAT and VAT; however, the chromatin accessibility landscape differences and how the cis-regulatory elements govern gene expression changes between SAT and VAT are unknown. Methods Pig were used to characterize the differences in chromatin accessibility between the two adipose depots-derived stromal vascular fractions (SVFs) using DNase-sequencing (DNase-seq). Using integrated data from DNase-seq, H3K27ac ChIP-sequencing (ChIP-seq), and RNA-sequencing (RNA-seq), we investigated how the regulatory locus complexity regulated gene expression changes between SAT and VAT and the possible impact that these changes may have on the different biological functions of these two adipose depots. Results SVFs form SAT and VAT (S-SVF and V-SVF) have differential chromatin accessibility landscapes. The differential DNase I hypersensitive site (DHS)-associated genes, which indicate dynamic chromatin accessibility, were mainly involved in metabolic processes and inflammatory responses. Additionally, the Krüppel-like factor family of transcription factors were enriched in the differential DHSs. Furthermore, the chromatin accessibility data were highly associated with differential gene expression as indicated using H3K27ac ChIP-seq and RNA-seq data, supporting the validity of the differential gene expression determined using DNase-seq. Moreover, by combining epigenetic and transcriptomic data, we identified two candidate genes, NR1D1 and CRYM, could be crucial to regulate distinct metabolic and inflammatory characteristics between SAT and VAT. Together, these results uncovered differences in the transcription regulatory network and enriched the mechanistic understanding of the different biological functions between SAT and VAT.
Collapse
Affiliation(s)
- Wenyang Sun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China,Key Laboratory of Pig Industry Sciences (Ministry of Agriculture), Chongqing Academy of Animal Science, Chongqing, China
| | - Tinghuan Zhang
- Key Laboratory of Pig Industry Sciences (Ministry of Agriculture), Chongqing Academy of Animal Science, Chongqing, China
| | - Silu Hu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qianzi Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xi Long
- Key Laboratory of Pig Industry Sciences (Ministry of Agriculture), Chongqing Academy of Animal Science, Chongqing, China
| | - Xu Yang
- College of Nursing, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Lei Chen
- Key Laboratory of Pig Industry Sciences (Ministry of Agriculture), Chongqing Academy of Animal Science, Chongqing, China
| |
Collapse
|
33
|
Liang J, Sun X, Yi L, Lv J. Effect of hyperbaric oxygen therapy on weight loss and hyperlipidemia in rats. Biochem Biophys Res Commun 2022; 599:106-112. [PMID: 35180469 DOI: 10.1016/j.bbrc.2022.02.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To explore whether hyperbaric oxygen therapy(HBO) can promote weight loss and recovery of hyperlipidemia in rats, and to explore the possible mechanism. METHODS 180 SD rats were divided into 6 groups with 30 rats in each group. The first 3 groups were intraperitoneally injected with fat emulsion to make hyperlipidemia models, and the last three groups were injected with normal saline. The first three groups received 3 h/d, 6 h/d, 0 h/d HBO therapy respectively, and the last three groups received the same treatment. Body weight, blood lipid and transaminase were measured in all SD rats, and pathological sections of heart, liver and kidney were observed. RESULTS Hyperlipidemia group treated with 3 h/d hyperbaric oxygen has the effect of reducing hyperlipidemia compared with other groups and has the effect of heart and kidney protection. Although 6 h/d HBO therapy has a more noticeable effect on lowering hyperlipidemia, it has more apparent liver damage effects. The normal group treated with HBO for 3 h/d or 6 h/d both have the effect of weight loss, and the impact of liver injury is not apparent. However, the 6 h/d HBO therapy group had a more prominent weight loss effect. CONCLUSION HBO therapy can promote weight loss and reduce hyperlipidemia. Our experiments have shown that 6 h/d and 3 h/d HBO therapy reduces blood lipids in hyperlipidemia SD rats. However, the former has noticeable liver damage effects on SD rats, and the latter is adequate for protecting the liver in normal or hyperlipidemia SD rats. At the same time, it has been proved that HBO therapy has cardio and kidney protection in hyperlipidemia SD rats.
Collapse
Affiliation(s)
- Jianhe Liang
- Department of Plastic Burn and Cosmetic Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, 154002, China
| | - Xu Sun
- Department of Burn, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lei Yi
- Department of Burn, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jinyuan Lv
- Department of Plastic Burn and Cosmetic Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, 154002, China.
| |
Collapse
|
34
|
Zapata J, Gallardo A, Romero C, Valenzuela R, Garcia-Diaz DF, Duarte L, Bustamante A, Gasaly N, Gotteland M, Echeverria F. n-3 polyunsaturated fatty acids in the regulation of adipose tissue browning and thermogenesis in obesity: Potential relationship with gut microbiota. Prostaglandins Leukot Essent Fatty Acids 2022; 177:102388. [PMID: 34995899 DOI: 10.1016/j.plefa.2021.102388] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 10/03/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Obesity is a worldwide public health problem characterized by fat tissue accumulation, favouring adipose tissue and metabolic alterations. Increasing energy expenditure (EE) through brown adipose tissue activation and white adipose tissue (WAT) browning has gained relevance as a therapeutic approach. Different bioactive compounds, such as n-3 polyunsaturated fatty acids (PUFA), have been shown to induce those thermogenic effects. This process is regulated by the gut microbiota as well. Nevertheless, obesity is characterized by gut microbiota dysbiosis, which can be restored by weight loss and n-3 PUFA intake, among other factors. Knowledge gap: However, the role of the gut microbiota on the n-3 PUFA effect in inducing thermogenesis in obesity has not been fully elucidated. OBJECTIVE This review aims to elucidate the potential implications of this interrelation on WAT browning adiposw sittue (BAT), BAT activity, and EE regulation in obesity models.
Collapse
Affiliation(s)
- J Zapata
- Escuela de Medicina, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - A Gallardo
- Escuela de Medicina, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - C Romero
- Escuela de Medicina, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - R Valenzuela
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Nutritional Sciences Department, Faculty of Medicine, University of Toronto, Toronto ON, Canada
| | - D F Garcia-Diaz
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - L Duarte
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - A Bustamante
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - N Gasaly
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile; ICBM: Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Facultad de Medicina, Universidad de Chile, Chile
| | - M Gotteland
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - F Echeverria
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Carrera de Nutricion y Dietetica, Departamento Ciencias de la Salud, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
35
|
Dani V, Yao X, Dani C. Transplantation of fat tissues and iPSC-derived energy expenditure adipocytes to counteract obesity-driven metabolic disorders: Current strategies and future perspectives. Rev Endocr Metab Disord 2022; 23:103-110. [PMID: 33751363 PMCID: PMC7982512 DOI: 10.1007/s11154-021-09632-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 12/25/2022]
Abstract
Several therapeutic options have been developed to address the obesity epidemic and treat associated metabolic diseases. Despite the beneficial effects of surgery and drugs, effective therapeutic solutions have been held back by the poor long-term efficiency and detrimental side effects. The development of alternative approaches is thus urgently required. Fat transplantation is common practice in many surgical procedures, including aesthetic and reconstructive surgery, and is a budding future direction for treating obesity-related metabolic defects. This review focuses on adipose tissue transplantation and the recent development of cell-based therapies to boost the mass of energy-expenditure cells. Brown adipocyte transplantation is a promising novel therapy to manage obesity and associated metabolic disorders, but the need to have an abundant and relevant source of brown fat tissue or brown adipocytes for transplantation is a major hurdle to overcome. Current studies have focused on the rodent model to obtain a proof of concept of a tissue-transplantation strategy able to achieve effective long-term effects to reverse metabolic defects in obese patients. Future perspectives and opportunities to develop innovative human fat tissue models and 3D engineered hiPSC-adipocytes are discussed.
Collapse
Affiliation(s)
- Vincent Dani
- Université Côte d'Azur, INSERM, CNRS, Nice, iBV, France
| | - Xi Yao
- Université Côte d'Azur, INSERM, CNRS, Nice, iBV, France
| | - Christian Dani
- Université Côte d'Azur, INSERM, CNRS, Nice, iBV, France.
| |
Collapse
|
36
|
Yonkova PY. Dynamics of the development of subcutaneous fat depots in rabbits – a gross anatomical and microscopic study. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2022. [DOI: 10.15547/bjvm.2373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In this study, gross anatomical and microscopic features of interscapular (IsFD) and inguinal (InFD) fat depots of 24 New Zealand White rabbits were evaluated. Rabbits were equally distributed into 4 groups: 1st - newborns, 2nd - 1 month old, 3rd - 2 months old and 4th - 3 months old. The cranial subcutaneous fat pad in newborns covered dorsal and ventral cervical and interscapular regions. As age advanced, cervical lobes underwent a rapid reduction but the development of interscapular lobes continued. IsFD in rabbits from 1st and 2nd group was composed of both white and brown adipocytes, while in 3rd and 4th groups it consisted of white adipocytes only. InFD in rabbits from all tested groups occupied respective inguinal region and no age-dependent changes in shape and topography were observed. In all groups InFD was composed of white adipocytes only. The highest growth rate of interscapular and inguinal adipocytes was established in one-month-old rabbits. Differences in anatomy and histology of interscapular and inguinal fat depots in rabbits could be successfully used for comparison in other experiments in the field of adipobiology and autologous transplantation, where fat depots undergo significant morphological changes.
Collapse
Affiliation(s)
- P. Y. Yonkova
- Department of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria
| |
Collapse
|
37
|
Milan G, Conci S, Sanna M, Favaretto F, Bettini S, Vettor R. ASCs and their role in obesity and metabolic diseases. Trends Endocrinol Metab 2021; 32:994-1006. [PMID: 34625375 DOI: 10.1016/j.tem.2021.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/23/2021] [Accepted: 09/03/2021] [Indexed: 01/04/2023]
Abstract
We describe adipose stromal/stem cells (ASCs) in the structural/functional context of the adipose tissue (AT) stem niche (adiponiche), including cell-cell interactions and the microenvironment, and emphasize findings obtained in humans and in lineage-tracing models. ASCs have distinctive markers, 'colors', and anatomical 'locations' which influence their functions. Each adiponiche component can become impaired, thereby contributing to the pathological AT alterations seen in obesity and metabolic diseases. We discuss adiposopathy with a focus on adiponiche dysfunction, and underline the mechanisms that control AT expansion and energy balance. Better understanding of adiponiche regulation and ASC features could help to identify therapeutic targets that favor weight loss and counteract weight regain, and also contribute to innovative strategies for regenerative medicine.
Collapse
Affiliation(s)
- Gabriella Milan
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy.
| | - Scilla Conci
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy
| | - Marta Sanna
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy
| | - Francesca Favaretto
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy
| | - Silvia Bettini
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy
| | - Roberto Vettor
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy
| |
Collapse
|
38
|
Lopez-Letayf S, Arie I, Araidy S, Abu El-Naaj I, Pitaru S, Arzate H. Human oral mucosa-derived neural crest-like stem cells differentiate into functional osteoprogenitors that contribute to regeneration of critical size calvaria defects. J Periodontal Res 2021; 57:305-315. [PMID: 34839539 DOI: 10.1111/jre.12960] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND OBJECTIVE Regeneration of large bony defects is an unmet medical need. The therapeutic effect of fully developed bony constructs engineered in vitro from mineralized scaffold and adult stem cells is hampered by deficient long-term graft integration. The purpose of the present study was to investigate the regenerative capacity of a bony primordial construct consisting of human oral mucosa stem cells (hOMSC)-derived osteoprogenitors and absorbable Gelfoam® sponges. METHODS Gingiva and alveolar mucosa-derived hOMSC were differentiated into osteoprogenitors (Runx2 and osterix positive) and loaded into Gelfoam® sponges to generate primordial hOMSC constructs. These were implanted into critical size calvaria defects in the rat. Defects treated with human dermal fibroblasts (HDF) constructs; Gelfoam® sponges and untreated defects served as controls. RESULTS After 120-day post-implantation defects treated with hOMSC constructs, HDF constructs and gelatin and untreated defects exhibited 86%, 30%, 21%, and 9% of new bone formation, respectively. Immunofluorescence analysis for human nuclear antigen (HNA), bone sialoprotein (BSP), and osteocalcin (OCN) revealed viable hOMSC-derived osteoblasts and osteocytes that formed most of the cell population of the newly formed bone at 30 and 120 days post surgery. Few HNA-positive HDF that were negative for BSP and OCN were identified together with inflammatory cells in the soft tissue adjacent to new bone formation only at 30 days post implantation. CONCLUSION Collectively, the results demonstrate that primordial in vitro engineered constructs consisting of hOMSC-derived osteoprogenitors and absorbable gelatin almost completely regenerate critical size defects in an immunocompetent xenogeneic animal by differentiating into functional osteoblasts that retain the immunomodulatory ability of naïve hOMSC.
Collapse
Affiliation(s)
- Sonia Lopez-Letayf
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Cd. Universitaria, Mexico City, Mexico
| | - Ina Arie
- Department of Oral Biology, School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shareef Araidy
- Department of Cranio-Maxillofacial Surgery, Baruch Padeh Medical Center, Poria, Israel
| | - Imad Abu El-Naaj
- Department of Cranio-Maxillofacial Surgery, Baruch Padeh Medical Center, Poria, Israel
| | - Sandu Pitaru
- Department of Oral Biology, School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Higinio Arzate
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Cd. Universitaria, Mexico City, Mexico
| |
Collapse
|
39
|
Tsagkaraki E, Nicoloro SM, DeSouza T, Solivan-Rivera J, Desai A, Lifshitz LM, Shen Y, Kelly M, Guilherme A, Henriques F, Amrani N, Ibraheim R, Rodriguez TC, Luk K, Maitland S, Friedline RH, Tauer L, Hu X, Kim JK, Wolfe SA, Sontheimer EJ, Corvera S, Czech MP. CRISPR-enhanced human adipocyte browning as cell therapy for metabolic disease. Nat Commun 2021; 12:6931. [PMID: 34836963 PMCID: PMC8626495 DOI: 10.1038/s41467-021-27190-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/08/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity and type 2 diabetes are associated with disturbances in insulin-regulated glucose and lipid fluxes and severe comorbidities including cardiovascular disease and steatohepatitis. Whole body metabolism is regulated by lipid-storing white adipocytes as well as "brown" and "brite/beige" adipocytes that express thermogenic uncoupling protein 1 (UCP1) and secrete factors favorable to metabolic health. Implantation of brown fat into obese mice improves glucose tolerance, but translation to humans has been stymied by low abundance of primary human beige adipocytes. Here we apply methods to greatly expand human adipocyte progenitors from small samples of human subcutaneous adipose tissue and then disrupt the thermogenic suppressor gene NRIP1 by CRISPR. Ribonucleoprotein consisting of Cas9 and sgRNA delivered ex vivo are fully degraded by the human cells following high efficiency NRIP1 depletion without detectable off-target editing. Implantation of such CRISPR-enhanced human or mouse brown-like adipocytes into high fat diet fed mice decreases adiposity and liver triglycerides while enhancing glucose tolerance compared to implantation with unmodified adipocytes. These findings advance a therapeutic strategy to improve metabolic homeostasis through CRISPR-based genetic enhancement of human adipocytes without exposing the recipient to immunogenic Cas9 or delivery vectors.
Collapse
Affiliation(s)
- Emmanouela Tsagkaraki
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- University of Crete School of Medicine, Crete, 71003, Greece
| | - Sarah M Nicoloro
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Tiffany DeSouza
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Javier Solivan-Rivera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Anand Desai
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Lawrence M Lifshitz
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Yuefei Shen
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Mark Kelly
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Adilson Guilherme
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Felipe Henriques
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Nadia Amrani
- University of Crete School of Medicine, Crete, 71003, Greece
| | - Raed Ibraheim
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Tomas C Rodriguez
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Kevin Luk
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Stacy Maitland
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Randall H Friedline
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Lauren Tauer
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Xiaodi Hu
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Erik J Sontheimer
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
40
|
Takaishi K, Oshima T, Eto H, Nishihira M, Nguyen ST, Ochi R, Fujita N, Urakawa S. Impact of Exercise and Detraining during Childhood on Brown Adipose Tissue Whitening in Obesity. Metabolites 2021; 11:metabo11100677. [PMID: 34677392 PMCID: PMC8540482 DOI: 10.3390/metabo11100677] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 11/25/2022] Open
Abstract
This study aimed to investigate the influence of childhood exercise and detraining on brown adipose tissue (BAT) whitening in obesity. Four-week-old male Long-Evans Tokushima Otsuka (LETO) rats (n = 9) and Otsuka Long-Evans Tokushima Fatty (OLETF) rats (n = 24) were used as non-obese and obese animals, respectively. OLETF rats were divided into non-exercise sedentary (n = 9) and exercise groups. OLETF rats in the exercise group were further divided into subgroups according to the exercise period—exercise from 10- to 12-weeks-old (n = 6); and exercise from 4- to 6-weeks-old, and detraining from 6- to 12-weeks-old (n = 9). At 12-weeks-old, immediately after exercise period, BAT whitening in OLETF rats was inhibited by exercise despite the fact that hypertrophy was not caused in the plantaris muscle. However, the effectiveness was attenuated during the detraining period. Histological BAT whitening and downregulation of uncoupling protein-1 (UCP-1) were found in non-exercise sedentary OLETF rats at 12-weeks-old. The downregulation was not inhibited, even though exercise histologically inhibited BAT whitening in OLETF rats. Childhood exercise decreased BAT whitening in obesity. Detraining attenuated the inhibition of BAT whitening. These results suggest that regular exercise is needed to improve BAT whitening and downregulation of UCP-1 in obesity.
Collapse
|
41
|
Sun JM, Ho CK, Gao Y, Chong CH, Zheng DN, Zhang YF, Yu L. Salvianolic acid-B improves fat graft survival by promoting proliferation and adipogenesis. Stem Cell Res Ther 2021; 12:507. [PMID: 34535194 PMCID: PMC8447755 DOI: 10.1186/s13287-021-02575-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022] Open
Abstract
Background Our previous study proved that Salvia miltiorrhiza could enhance fat graft survival by promoting adipogenesis. However, the effect of salvianolic acid B (Sal-B), the most abundant and bioactive water-soluble compound in Salvia miltiorrhiza, on fat graft survival has not yet been investigated. Objective This study aims to investigate whether salvianolic acid B could improve fat graft survival and promote preadipocyte differentiation. The underlying mechanism has also been studied. Methods In vivo, 0.2 ml of Coleman fat was transplanted into nude mice with salvianolic acid B. The grafts were evaluated by HE and IF at 2 and 4 weeks posttransplantation and by micro-CT at 4 weeks posttransplantation. In vitro, the adipogenesis and proliferative activities of salvianolic acid B were analyzed in cultured human adipose-derived stem cells (h-ADSCs) and 3T3-L1 cells to detect the mechanism by which salvianolic acid B affects graft survival. Results In vivo, the weights and volumes of the fat grafts in the Sal-B-treated groups were significantly higher than those of the fat grafts in the control group. In addition, higher fat integrity and more viable adipocytes were observed in the Sal-B-treated groups. In vitro, salvianolic acid B showed the ability to promote 3T3-L1 and h-ADSC proliferation and adipogenesis. Conclusions Our in vitro experiments demonstrated that salvianolic acid B can promote the proliferation of adipose stem cells and enhance the differentiation of adipose stem cells. Simultaneously, in vivo experiments showed that salvianolic acid B can improve the survival rate of fat transplantation. Therefore, our research shed light on the potential therapeutic usage of salvianolic acid B in improving the survival rate of fat transplantation. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02575-4.
Collapse
Affiliation(s)
- Jia-Ming Sun
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhi Zao Ju Road, Shanghai, People's Republic of China, 200011
| | - Chia-Kang Ho
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhi Zao Ju Road, Shanghai, People's Republic of China, 200011
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhi Zao Ju Road, Shanghai, People's Republic of China, 200011
| | - Chio-Hou Chong
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhi Zao Ju Road, Shanghai, People's Republic of China, 200011
| | - Dan-Ning Zheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhi Zao Ju Road, Shanghai, People's Republic of China, 200011.
| | - Yi-Fan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhi Zao Ju Road, Shanghai, People's Republic of China, 200011.
| | - Li Yu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhi Zao Ju Road, Shanghai, People's Republic of China, 200011.
| |
Collapse
|
42
|
Sharif O, Brunner JS, Korosec A, Martins R, Jais A, Snijder B, Vogel A, Caldera M, Hladik A, Lakovits K, Saluzzo S, Boehm B, Gorki AD, Mesteri I, Lindroos-Christensen J, Tillmann K, Stoiber D, Menche J, Schabbauer G, Bilban M, Superti-Furga G, Esterbauer H, Knapp S. Beneficial Metabolic Effects of TREM2 in Obesity Are Uncoupled From Its Expression on Macrophages. Diabetes 2021; 70:2042-2057. [PMID: 33627323 PMCID: PMC8576425 DOI: 10.2337/db20-0572] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 02/19/2021] [Indexed: 12/03/2022]
Abstract
Obesity-induced white adipose tissue (WAT) hypertrophy is associated with elevated adipose tissue macrophage (ATM) content. Overexpression of the triggering receptor expressed on myeloid cells 2 (TREM2) reportedly increases adiposity, worsening health. Paradoxically, using insulin resistance, elevated fat mass, and hypercholesterolemia as hallmarks of unhealthy obesity, a recent report demonstrated that ATM-expressed TREM2 promoted health. Here, we identified that in mice, TREM2 deficiency aggravated diet-induced insulin resistance and hepatic steatosis independently of fat and cholesterol levels. Metabolomics linked TREM2 deficiency with elevated obesity-instigated serum ceramides that correlated with impaired insulin sensitivity. Remarkably, while inhibiting ceramide synthesis exerted no influences on TREM2-dependent ATM remodeling, inflammation, or lipid load, it restored insulin tolerance, reversing adipose hypertrophy and secondary hepatic steatosis of TREM2-deficient animals. Bone marrow transplantation experiments revealed unremarkable influences of immune cell-expressed TREM2 on health, instead demonstrating that WAT-intrinsic mechanisms impinging on sphingolipid metabolism dominate in the systemic protective effects of TREM2 on metabolic health.
Collapse
Affiliation(s)
- Omar Sharif
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria
- Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Julia Stefanie Brunner
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria
- Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Ana Korosec
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Rui Martins
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Alexander Jais
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Berend Snijder
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Andrea Vogel
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria
- Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Michael Caldera
- Department of Structural and Computational Biology, Max Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Anastasiya Hladik
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Karin Lakovits
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Simona Saluzzo
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Benedikta Boehm
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Anna-Dorothea Gorki
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | | | - Katharina Tillmann
- Center of Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Dagmar Stoiber
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Division of Pharmacology, Department of Pharmacology, Physiology and Microbiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Jörg Menche
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Structural and Computational Biology, Max Perutz Laboratories, University of Vienna, Vienna, Austria
- Faculty of Mathematics, University of Vienna, Vienna, Austria
| | - Gernot Schabbauer
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria
- Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Giulio Superti-Furga
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Sylvia Knapp
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| |
Collapse
|
43
|
Yang J, Zhang H, Parhat K, Xu H, Li M, Wang X, Ran C. Molecular Imaging of Brown Adipose Tissue Mass. Int J Mol Sci 2021; 22:ijms22179436. [PMID: 34502347 PMCID: PMC8431742 DOI: 10.3390/ijms22179436] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 12/28/2022] Open
Abstract
Brown adipose tissue (BAT), a uniquely thermogenic tissue that plays an important role in metabolism and energy expenditure, has recently become a revived target in the fight against metabolic diseases, such as obesity, diabetes, and non-alcoholic fatty liver disease (NAFLD). Different from white adipose tissue (WAT), the brown adipocytes have distinctive features including multilocular lipid droplets, a large number of mitochondria, and a high expression of uncoupling protein-1 (UCP-1), as well as abundant capillarity. These histologic characteristics provide an opportunity to differentiate BAT from WAT using imaging modalities, such as PET/CT, SPECT/CT, MRI, NIRF and Ultrasound. However, most of the reported imaging methods were BAT activation dependent, and the imaging signals could be affected by many factors, including environmental temperatures and the states of the sympathetic nervous system. Accurate BAT mass detection methods that are independent of temperature and hormone levels have the capacity to track the development and changes of BAT throughout the lifetime of mammals, and such methods could be very useful for the investigation of potential BAT-related therapies. In this review, we focus on molecular imaging modalities that can detect and quantify BAT mass. In addition, their detection mechanism and limitations will be discussed as well.
Collapse
Affiliation(s)
- Jing Yang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (K.P.); (H.X.); (M.L.); (X.W.)
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, MA 02129, USA
- Correspondence: (J.Y.); (C.R.)
| | - Haili Zhang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (K.P.); (H.X.); (M.L.); (X.W.)
| | - Kadirya Parhat
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (K.P.); (H.X.); (M.L.); (X.W.)
| | - Hui Xu
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (K.P.); (H.X.); (M.L.); (X.W.)
| | - Mingshuang Li
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (K.P.); (H.X.); (M.L.); (X.W.)
| | - Xiangyu Wang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China; (H.Z.); (K.P.); (H.X.); (M.L.); (X.W.)
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, MA 02129, USA
- Correspondence: (J.Y.); (C.R.)
| |
Collapse
|
44
|
Nogueiras R. MECHANISMS IN ENDOCRINOLOGY: The gut-brain axis: regulating energy balance independent of food intake. Eur J Endocrinol 2021; 185:R75-R91. [PMID: 34260412 PMCID: PMC8345901 DOI: 10.1530/eje-21-0277] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
Obesity is a global pandemic with a large health and economic burden worldwide. Bodyweight is regulated by the ability of the CNS, and especially the hypothalamus, to orchestrate the function of peripheral organs that play a key role in metabolism. Gut hormones play a fundamental role in the regulation of energy balance, as they modulate not only feeding behavior but also energy expenditure and nutrient partitioning. This review examines the recent discoveries about hormones produced in the stomach and gut, which have been reported to regulate food intake and energy expenditure in preclinical models. Some of these hormones act on the hypothalamus to modulate thermogenesis and adiposity in a food intake-independent fashion. Finally, the association of these gut hormones to eating, energy expenditure, and weight loss after bariatric surgery in humans is discussed.
Collapse
Affiliation(s)
- Ruben Nogueiras
- Department of Physiology, CIMUS, USC, CIBER Fisiopatología Obesidad y Nutrición (CiberOBN), Instituto Salud Carlos III, Galician Agency of Innovation, Xunta de Galicia, Santiago de Compostela, Spain
| |
Collapse
|
45
|
Sakane S, Hikita H, Shirai K, Myojin Y, Sasaki Y, Kudo S, Fukumoto K, Mizutani N, Tahata Y, Makino Y, Yamada R, Kodama T, Sakamori R, Tatsumi T, Takehara T. White Adipose Tissue Autophagy and Adipose-Liver Crosstalk Exacerbate Nonalcoholic Fatty Liver Disease in Mice. Cell Mol Gastroenterol Hepatol 2021; 12:1683-1699. [PMID: 34303881 PMCID: PMC8551788 DOI: 10.1016/j.jcmgh.2021.07.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Although nonalcoholic fatty liver disease (NAFLD) is closely associated with obesity, the role of adipose tissue in NAFLD is not well-understood. Because autophagy has been reported to be involved in the degradation of lipid droplets, we investigated the role of adipose tissue autophagy in the liver pathogenesis of NAFLD. METHODS C57BL/6J mice and adipocyte-specific Atg7-knockout mice (Adipoq-Atg7 KO mice) were fed a high-fat diet (HFD). RESULTS HFD feeding for up to 4 months increased both inguinal and epididymal white adipose tissue (iWAT and eWAT, respectively; the former represents subcutaneous fat, and the latter represents visceral fat) in mice. After HFD feeding, autophagy flux in both types of white adipose tissue was increased, and the levels of Rubicon, a negative autophagy regulator, were decreased, suggesting autophagy promotion. Adipoq-Atg7 KO mice exhibited suppressed autophagy in both iWAT and eWAT. Adipocyte-specific Atg7 KO enhanced HFD-induced iWAT hypertrophy. On the other hand, eWAT levels in Adipoq-Atg7 KO mice were increased after 1 month of HFD feeding but decreased after 4 months of HFD feeding compared with those in wild-type controls. Cleaved caspase 3 and JNK pathway protein expression in eWAT was increased without cytokine elevation in Adipoq-Atg7 KO mice fed an HFD compared with wild-type mice fed an HFD. Adipocyte-specific Atg7 KO decreased serum free fatty acid levels and ameliorated HFD-induced steatosis, liver inflammation, and fibrosis. CONCLUSIONS Autophagy was enhanced in the white adipose tissues of mice fed an HFD. Autophagy inhibition in white adipose tissues ameliorated the liver pathology of NAFLD via adipose-liver crosstalk.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Tetsuo Takehara
- Correspondence Address correspondence to: Tetsuo Takehara, MD, PhD, 2-2 Yamadaoka, Suita, Osaka, 565-0871 Japan. fax: +81-6-6879-3629.
| |
Collapse
|
46
|
King BC, Blom AM. Complement in metabolic disease: metaflammation and a two-edged sword. Semin Immunopathol 2021; 43:829-841. [PMID: 34159399 PMCID: PMC8613079 DOI: 10.1007/s00281-021-00873-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/23/2021] [Indexed: 01/28/2023]
Abstract
We are currently experiencing an enduring global epidemic of obesity and diabetes. It is now understood that chronic low-grade tissue inflammation plays an important role in metabolic disease, brought upon by increased uptake of a so-called Western diet, and a more sedentary lifestyle. Many evolutionarily conserved links exist between metabolism and the immune system, and an imbalance in this system induced by chronic over-nutrition has been termed 'metaflammation'. The complement system is an important and evolutionarily ancient part of innate immunity, but recent work has revealed that complement not only is involved in the recognition of pathogens and induction of inflammation, but also plays important roles in cellular and tissue homeostasis. Complement can therefore contribute both positively and negatively to metabolic control, depending on the nature and anatomical site of its activity. This review will therefore focus on the interactions of complement with mechanisms and tissues relevant for metabolic control, obesity and diabetes.
Collapse
Affiliation(s)
- B C King
- Department of Translational Medicine, Lund University, Lund, Sweden.
| | - A M Blom
- Department of Translational Medicine, Lund University, Lund, Sweden
| |
Collapse
|
47
|
Cartee GD. Sexual Dimorphic Effects of Exercise Training on Subcutaneous White Adipose Tissue of Mice. Diabetes 2021; 70:1242-1243. [PMID: 34016599 PMCID: PMC8275887 DOI: 10.2337/dbi21-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology; Department of Molecular and Integrative Physiology; and Institute of Gerontology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
48
|
Brandão BB, Poojari A, Rabiee A. Thermogenic Fat: Development, Physiological Function, and Therapeutic Potential. Int J Mol Sci 2021; 22:5906. [PMID: 34072788 PMCID: PMC8198523 DOI: 10.3390/ijms22115906] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
The concerning worldwide increase of obesity and chronic metabolic diseases, such as T2D, dyslipidemia, and cardiovascular disease, motivates further investigations into preventive and alternative therapeutic approaches. Over the past decade, there has been growing evidence that the formation and activation of thermogenic adipocytes (brown and beige) may serve as therapy to treat obesity and its associated diseases owing to its capacity to increase energy expenditure and to modulate circulating lipids and glucose levels. Thus, understanding the molecular mechanism of brown and beige adipocytes formation and activation will facilitate the development of strategies to combat metabolic disorders. Here, we provide a comprehensive overview of pathways and players involved in the development of brown and beige fat, as well as the role of thermogenic adipocytes in energy homeostasis and metabolism. Furthermore, we discuss the alterations in brown and beige adipose tissue function during obesity and explore the therapeutic potential of thermogenic activation to treat metabolic syndrome.
Collapse
Affiliation(s)
- Bruna B. Brandão
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Ankita Poojari
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| | - Atefeh Rabiee
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| |
Collapse
|
49
|
Porro S, Genchi VA, Cignarelli A, Natalicchio A, Laviola L, Giorgino F, Perrini S. Dysmetabolic adipose tissue in obesity: morphological and functional characteristics of adipose stem cells and mature adipocytes in healthy and unhealthy obese subjects. J Endocrinol Invest 2021; 44:921-941. [PMID: 33145726 DOI: 10.1007/s40618-020-01446-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
The way by which subcutaneous adipose tissue (SAT) expands and undergoes remodeling by storing excess lipids through expansion of adipocytes (hypertrophy) or recruitment of new precursor cells (hyperplasia) impacts the risk of developing cardiometabolic and respiratory diseases. In unhealthy obese subjects, insulin resistance, type 2 diabetes, hypertension, and obstructive sleep apnoea are typically associated with pathologic SAT remodeling characterized by adipocyte hypertrophy, as well as chronic inflammation, hypoxia, increased visceral adipose tissue (VAT), and fatty liver. In contrast, metabolically healthy obese individuals are generally associated with SAT development characterized by the presence of smaller and numerous mature adipocytes, and a lower degree of VAT inflammation and ectopic fat accumulation. The remodeling of SAT and VAT is under genetic regulation and influenced by inherent depot-specific differences of adipose tissue-derived stem cells (ASCs). ASCs have multiple functions such as cell renewal, adipogenic capacity, and angiogenic properties, and secrete a variety of bioactive molecules involved in vascular and extracellular matrix remodeling. Understanding the mechanisms regulating the proliferative and adipogenic capacity of ASCs from SAT and VAT in response to excess calorie intake has become a focus of interest over recent decades. Here, we summarize current knowledge about the biological mechanisms able to foster or impair the recruitment and adipogenic differentiation of ASCs during SAT and VAT development, which regulate body fat distribution and favorable or unfavorable metabolic responses.
Collapse
Affiliation(s)
- S Porro
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - V A Genchi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Cignarelli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Natalicchio
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - L Laviola
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - F Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy.
| | - S Perrini
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
50
|
Tan L, Tran L, Ferreyra S, Moran JA, Skovgaard Z, Trujillo A, ibili E, Zhao Y. Downregulation of SUV39H1 and CITED2 Exerts Additive Effect on Promoting Adipogenic Commitment of Human Mesenchymal Stem Cells. Stem Cells Dev 2021; 30:485-501. [PMID: 33691475 PMCID: PMC8106253 DOI: 10.1089/scd.2020.0190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/10/2021] [Indexed: 11/12/2022] Open
Abstract
Human adipogenesis is the process through which uncommitted human mesenchymal stem cells (hMSCs) differentiate into adipocytes. Through a siRNA-based high-throughput screen that identifies adipogenic regulators whose expression knockdown leads to enhanced adipogenic differentiation of hMSCs, two new regulators, SUV39H1, a histone methyltransferase that catalyzes H3K9Me3, and CITED2, a CBP/p300-interacting transactivator with Glu/Asp-rich carboxy-terminal domain 2 were uncovered. Both SUV39H1 and CITED2 are normally downregulated during adipogenic differentiation of hMSCs. Further expression knockdown induced by siSUV39H1 or siCITED2 at the adipogenic initiation stage significantly enhanced adipogenic differentiation of hMSCs as compared with siControl treatment, with siSUV39H1 acting by both accelerating fat accumulation in individual adipocytes and increasing the total number of committed adipocytes, whereas siCITED2 acting predominantly by increasing the total number of committed adipocytes. In addition, both siSUV39H1 and siCITED2 were able to redirect hMSCs to undergo adipogenic differentiation in the presence of osteogenic inducing media, which normally only induces osteogenic differentiation of hMSCs in the absence of siSUV39H1 or siCITED2. Interestingly, simultaneous knockdown of both SUV39H1 and CITED2 resulted in even greater levels of adipogenic differentiation of hMSCs and expression of CEBPα and PPARγ, two master regulators of adipogenesis, as compared with those elicited by single gene knockdown. Furthermore, the effects of co-knockdown were equivalent to the additive effect of individual gene knockdown. Taken together, this study demonstrates that SUV39H1 and CITED2 are both negative regulators of human adipogenesis, and downregulation of both genes exerts an additive effect on promoting adipogenic differentiation of hMSCs through augmented commitment.
Collapse
Affiliation(s)
- Lun Tan
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Linh Tran
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Stephanie Ferreyra
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Jose A. Moran
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Zachary Skovgaard
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Amparo Trujillo
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Esra ibili
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Yuanxiang Zhao
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| |
Collapse
|