1
|
Sheikh E, Liu Q, Burk D, Beavers WN, Fu X, Gartia MR. Mapping lipid species remodeling in high fat diet-fed mice: Unveiling adipose tissue dysfunction with Raman microspectroscopy. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159557. [PMID: 39128539 PMCID: PMC11380576 DOI: 10.1016/j.bbalip.2024.159557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Dysregulated lipid metabolism in obesity leads to adipose tissue expansion, a major contributor to metabolic dysfunction and chronic disease. Lipid metabolism and fatty acid changes play vital roles in the progression of obesity. In this proof-of-concept study, Raman techniques combined with histochemical imaging methods were utilized to analyze the impact of a high-fat diet (HFD) on different types of adipose tissue in mice, using a small sample size (n = 3 per group). After six weeks of high-fat diet (HFD) feeding, our findings showed hypertrophy, elevated collagen levels, and increased macrophage presence in the adipose tissues of the HFD group compared to the low-fat diet (LFD) group. Statistical analysis of Raman spectra revealed significantly lower unsaturated lipid levels and higher lipid to protein content in different fat pads (brown adipose tissue (BAT), subcutaneous white adipose tissue (SWAT), and visceral white adipose tissue (VWAT)) with HFD. Raman images of adipose tissues were analyzed using Empty modeling and DCLS methods to spatially profile unsaturated and saturated lipid species in the tissues. It revealed elevated levels of ω-3, ω-6, cholesterol, and triacylglycerols in BAT adipose tissues of HFD compared to LFD tissues. These findings indicated that while cholesterol, ω-6/ω-3 ratio, and triacylglycerol levels have risen in the SWAT and VWAT adipose tissues of the HFD group, the levels of ω-3 and ω-6 have decreased following the HFD. The study showed that Raman spectroscopy provided invaluable information at the molecular level for investigating lipid species remodeling and spatial mapping of adipose tissues during HFD.
Collapse
Affiliation(s)
- Elnaz Sheikh
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Qianglin Liu
- LSU AgCenter, School of Animal Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - David Burk
- Cell Biology and Bioimaging Core, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - William N Beavers
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; Louisiana State University School of Veterinary Medicine, Mass Spectrometry Resource Center, Baton Rouge, LA 70803, USA
| | - Xing Fu
- LSU AgCenter, School of Animal Sciences, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
2
|
Wang J, Xie F, Zhu W, Ye D, Xiao Y, Shi M, Zeng R, Bian J, Xu X, Chen L, Zhu A, Zhu K, Fan T, Liu B, Xiao L, Zhang X. Relationship between serum carotenoids and telomere length in overweight or obese individuals. Front Nutr 2024; 11:1479994. [PMID: 39650708 PMCID: PMC11620882 DOI: 10.3389/fnut.2024.1479994] [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: 08/13/2024] [Accepted: 11/13/2024] [Indexed: 12/11/2024] Open
Abstract
Background Previous researches have demonstrated an association between carotenoids and elongated telomeres. Nonetheless, there is scant scientific evidence examining this relationship in individuals who are overweight or obese, a demographic more predisposed to accelerated aging. This study aims to elucidate the correlation between serum carotenoid concentrations and telomere length within this population group. Methods Data were sourced from the 2001-2002 National Health and Nutrition Examination Survey, encompassing 2,353 overweight or obese participants. The levels of α-carotene, β-carotene (both trans and cis isomers), β-cryptoxanthin, lutein/zeaxanthin, and trans-lycopene were quantified via high-performance liquid chromatography. Telomere length was assessed using quantitative polymerase chain reaction. Results Following adjustment for potential confounders, telomere length exhibited an increase of 1.83 base pairs (bp) per unit elevation in β-carotene levels (β = 1.83; 95% CI: 0.48, 3.18). Within the fully adjusted model, telomere length incremented by 1.7 bp per unit increase in serum β-carotene among overweight individuals (β = 1.7; 95% CI: 0.1, 3.3), and by 2.6 bp per unit increase among obese individuals (β = 2.6; 95% CI: 0.1, 5.0). Furthermore, restricted cubic spline analysis revealed a linear relationship between β-carotene levels and telomere length, whereas a non-linear association was observed between β-cryptoxanthin levels and telomere length. Conclusion This investigation indicates that higher serum β-carotene concentrations are linked with extended telomere length in overweight and obese populations in the United States. These findings warrant further validation through prospective studies.
Collapse
Affiliation(s)
- Jiang Wang
- School of Basic Medicine, Jinggangshan University, Ji’an, China
- Online Collaborative Research Center for Evidence-Based Medicine Ministry of Education, Jinggangshan University, Ji’an, China
| | - Fayi Xie
- School of Clinical Medicine, Jinggangshan University, Ji’an, China
| | - Wan Zhu
- School of Clinical Medicine, Jinggangshan University, Ji’an, China
| | - Dongmei Ye
- School of Clinical Medicine, Jinggangshan University, Ji’an, China
| | - Yi Xiao
- School of Mathematics and Physics, Jinggangshan University, Ji’an, China
| | - Mengxia Shi
- School of Clinical Medicine, Jinggangshan University, Ji’an, China
| | - Rui Zeng
- School of Clinical Medicine, Jinggangshan University, Ji’an, China
| | - Jiahui Bian
- School of Clinical Medicine, Jinggangshan University, Ji’an, China
| | - Xiao Xu
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lihuan Chen
- School of Chinese Medicine, Jinggangshan University, Ji’an, China
| | - Aizhang Zhu
- School of Basic Medicine, Jinggangshan University, Ji’an, China
| | - Ke Zhu
- School of Clinical Medicine, Jinggangshan University, Ji’an, China
| | - Tenghui Fan
- School of Clinical Medicine, Jinggangshan University, Ji’an, China
| | - Bin Liu
- School of Basic Medicine, Jinggangshan University, Ji’an, China
| | - Liyan Xiao
- School of Foreign Languages, Jinggangshan University, Ji’an, China
| | - Xiaoming Zhang
- Department of Emergency, The People’s Hospital of Baoan Shenzhen, Shenzhen, China
| |
Collapse
|
3
|
Pomar CA, Trepiana J, Besné-Eseverri I, Castillo P, Palou A, Palou M, Portillo MP, Picó C. Maternal Dietary Improvement or Leptin Supplementation During Suckling Mitigates the Long-Term Impact of Maternal Obesogenic Conditions on Inflammatory and Oxidative Stress Biomarkers in the Offspring of Diet-Induced Obese Rats. Int J Mol Sci 2024; 25:11876. [PMID: 39595945 PMCID: PMC11594198 DOI: 10.3390/ijms252211876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
This study investigates the impact of maternal nutrition during lactation on inflammation and oxidative stress in the offspring of diet-induced obese rats, along with the potential benefits of leptin supplementation during suckling. Dams were fed either a standard diet (SD), a western diet (WD) before and during gestation and lactation (WD-dams), or a WD switched to an SD during lactation (Rev-dams). Offspring were supplemented with leptin or vehicle during suckling and then fed an SD or WD until four months. Offspring of the Rev-dams exhibited improved metabolic indicators, including lower body weight, reduced plasma levels of TNF-alpha, a higher adiponectin/leptin (A/L) ratio, enhanced liver antioxidant defenses, and decreased inflammation markers in white adipose tissue (WAT) compared to WD-dams, with sex differences. Leptin supplementation further modulated these markers, reducing oxidative stress in liver and inflammation in WAT and liver (e.g., hepatic Tnfa expression decreased by 45% (males) and 41% (females) in the WD group on an SD), and improving the A/L ratio, with effects varying by maternal conditions and sex. In conclusion, this study underscores the importance of maternal nutrition and leptin intake during suckling in shaping long-term metabolic and inflammatory health in offspring, offering strategies to mitigate the adverse effects of maternal obesity on future generations.
Collapse
Affiliation(s)
- Catalina Amadora Pomar
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (P.C.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
| | - Jenifer Trepiana
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, Lucio Lascaray Research Centre, University of the Basque Country (UPV/EHU), 01006 Vitoria, Spain
- BIOARABA Institute of Health, 01006 Vitoria-Gasteiz, Spain
| | - Irene Besné-Eseverri
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, Lucio Lascaray Research Centre, University of the Basque Country (UPV/EHU), 01006 Vitoria, Spain
| | - Pedro Castillo
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (P.C.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (P.C.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), 07122 Palma, Spain
| | - Mariona Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (P.C.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
| | - Maria P. Portillo
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, Lucio Lascaray Research Centre, University of the Basque Country (UPV/EHU), 01006 Vitoria, Spain
- BIOARABA Institute of Health, 01006 Vitoria-Gasteiz, Spain
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (P.C.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), 07122 Palma, Spain
| |
Collapse
|
4
|
Hu J, Deng X, Li L, Liu H, Tang F, Ding Z, Guo B, Fan Y, Lu L. The Predictive Factors of Combined Implant Application for Breast Cancer Patients Receiving Immediate Breast Reconstruction with a Pedicled Omental Flap. Ann Surg Oncol 2024; 31:8362-8371. [PMID: 39048897 DOI: 10.1245/s10434-024-15882-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Whether a laparoscopically harvested omental flap is adequate for total breast reconstruction could not be determined preoperativaly due to lack of reliable assessment methods. This study aimed to establish a statistical model to predict the probability of omental flap insufficiency. METHODS In this study, 200 female patients with breast cancer receiving immediate breast reconstruction with pure pedicled omental flaps or pedicled omental flaps combined with implants after nipple-areolar complex-sparing mastectomy were divided into two groups depending on whether implants were needed or not. The clinical characteristics of these two groups were compared. Correlation of body mass index (BMI) and omental volume was analyzed. Binary logistic regression was performed to predict the probability of implant requirement based on clinical parameters, showing significant differences between the two groups. RESULTS The patients who needed implants in adjunct treatment were younger. In addition, they had larger breast specimens and smaller omental volumes than the others whose omental flaps were sufficient for total breast reconstruction. Body mass index and omental volume showed a moderately positive correlation. Age, specimen volume, and BMI all were entered into the logistic regression equation. For the patients with a BMI lower than 24.0 kg/m2, the probability of requiring implants was 5.467 times that of comparable patients with a BMI of 24.0 kg/m2 or higher. At the cutoff of 0.61, the regression equation yielded a sensitivity of 84.2% and a specificity of 72.1% in recognizing subjects with the necessity of implant application. CONCLUSION The combination of BMI, age, and volume of breast specimen could predict with high accuracy whether implants are required for breast cancer patients receiving pedicled omental flap-based breast reconstruction.
Collapse
Affiliation(s)
- Junda Hu
- Department of Thyroid, Breast, and Hernia Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xianzhao Deng
- Department of Thyroid, Breast, and Hernia Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Li
- The Second Department of Breast Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hao Liu
- The Second Department of Breast Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Fen Tang
- Department of Thyroid, Breast, and Hernia Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Ding
- Department of Thyroid, Breast, and Hernia Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bomin Guo
- Department of Thyroid, Breast, and Hernia Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youben Fan
- Department of Thyroid, Breast, and Hernia Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingli Lu
- Department of Thyroid, Breast, and Hernia Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Contessi Negrini N, Pellegrinelli V, Salem V, Celiz A, Vidal-Puig A. Breaking barriers in obesity research: 3D models of dysfunctional adipose tissue. Trends Biotechnol 2024:S0167-7799(24)00278-6. [PMID: 39443224 DOI: 10.1016/j.tibtech.2024.09.017] [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: 07/27/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Obesity is a global health crisis characterised by excessive accumulation of adipose tissue (AT). Under obesogenic conditions, this metabolically active tissue undergoes fibrosis and inflammation, leading to obesity-linked comorbidities. Modelling AT is essential for understanding its pathophysiology and developing treatments to protect against metabolic complications. 3D in vitro AT models are promising tools that address the limitations of traditional 2D in vitro models and in vivo animal models, providing enhanced biomimetic and human-relevant platforms. 3D models facilitate the study of AT pathophysiology and therapeutic screening. This review discusses the crucial role of AT in obesity-linked comorbidities, its dynamicity and complexity, and recent advances in engineering 3D scaffold-based in vitro dysfunctional AT models, highlighting potential breakthroughs in metabolic research and beyond.
Collapse
Affiliation(s)
- Nicola Contessi Negrini
- Department of Bioengineering, Imperial College London, London, UK; The Francis Crick Institute, London, UK.
| | | | - Victoria Salem
- Department of Bioengineering, Imperial College London, London, UK
| | - Adam Celiz
- Department of Bioengineering, Imperial College London, London, UK; The Francis Crick Institute, London, UK
| | - Antonio Vidal-Puig
- MRC Institute of Metabolic Science and Medical Research Council, Cambridge, UK; Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, PR China; Centro de Investigacion Principe Felipe (CIPF), Valencia, Spain; Cambridge Heart and Lung Research Institute, Cambridge, UK
| |
Collapse
|
6
|
Brito ML, Coutinho-Wolino KS, Almeida PP, Trigueira PDC, Alves APDP, Magliano DC, Stockler-Pinto MB. Unstressing the Reticulum: Nutritional Strategies for Modulating Endoplasmic Reticulum Stress in Obesity. Mol Nutr Food Res 2024; 68:e2400361. [PMID: 39363792 DOI: 10.1002/mnfr.202400361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/03/2024] [Indexed: 10/05/2024]
Abstract
The progression of obesity involves several molecular mechanisms that are closely associated with the pathophysiological response of the disease. Endoplasmic reticulum (ER) stress is one such factor. Lipotoxicity disrupts endoplasmic reticulum homeostasis in the context of obesity. Furthermore, it induces ER stress by activating several signaling pathways via inflammatory responses and oxidative stress. ER performs crucial functions in protein synthesis and lipid metabolism; thus, triggers such as lipotoxicity can promote the accumulation of misfolded proteins in the organelle. The accumulation of these proteins can lead to metabolic disorders and chronic inflammation, resulting in cell death. Thus, alternatives, such as flavonoids, amino acids, and polyphenols that are associated with antioxidant and anti-inflammatory responses have been proposed to attenuate this response by modulating ER stress via the administration of nutrients and bioactive compounds. Decreasing inflammation and oxidative stress can reduce the expression of several ER stress markers and improve clinical outcomes through the management of obesity, including the control of body weight, visceral fat, and lipid accumulation. This review explores the metabolic changes resulting from ER stress and discusses the role of nutritional interventions in modulating the ER stress pathway in obesity.
Collapse
Affiliation(s)
- Michele Lima Brito
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | - Karen Salve Coutinho-Wolino
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | - Patricia Pereira Almeida
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | | | - Ana Paula de Paula Alves
- Endocrinology Post Graduate Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 24210-201, Brazil
| | - D'Angelo Carlo Magliano
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Endocrinology Post Graduate Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 24210-201, Brazil
- Morphology Department, Biomedical Institute, Fluminense Federal University (UFF), Niterói, RJ, 24020-150, Brazil
| | - Milena Barcza Stockler-Pinto
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Nutrition Sciences Postgraduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24020-140, Brazil
| |
Collapse
|
7
|
Jack BU, Dias S, Pheiffer C. Comparative Effects of Tumor Necrosis Factor Alpha, Lipopolysaccharide, and Palmitate on Mitochondrial Dysfunction in Cultured 3T3-L1 Adipocytes. Cell Biochem Biophys 2024:10.1007/s12013-024-01522-3. [PMID: 39269560 DOI: 10.1007/s12013-024-01522-3] [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] [Accepted: 08/29/2024] [Indexed: 09/15/2024]
Abstract
We have previously reported that dysregulated lipid metabolism and inflammation in 3T3-L1 adipocytes is attributed to tumor necrosis factor alpha (TNFα) rather than lipopolysaccharide (LPS) and palmitate (PA). In this study, we further compared the modulative effects of TNFα, LPS, and PA on mitochondrial function by treating 3T3-L1 adipocytes with TNFα (10 ng/mL), LPS (100 ng/mL), and PA (0.75 mM) individually or in combination for 24 h. Results showed a significant reduction in intracellular adenosine triphosphate (ATP) content, mitochondrial bioenergetics, total antioxidant capacity, and the mRNA expression of citrate synthase (Cs), sirtuin 3 (Sirt3), protein kinase AMP-activated catalytic subunit alpha 2 (Prkaa2), peroxisome proliferator-activated receptor gamma coactivator 1 alpha (Ppargc1α), nuclear respiratory factor 1 (Nrf1), and superoxide dismutase 1 (Sod1) in cells treated with TNFα individually or in combination with LPS and PA. Additionally, TNFα treatments decreased insulin receptor substrate 1 (Irs1), insulin receptor substrate 2 (Irs2), solute carrier family 2, facilitated glucose transporter member 4 (Slc2a4), and phosphoinositide 3 kinase regulatory subunit 1 (Pik3r1) mRNA expression. Treatment with LPS and PA alone, or in combination, did not affect the assessed metabolic parameters, while the combination of LPS and PA increased lipid peroxidation. These results show that TNFα but not LPS and PA dysregulate mitochondrial function, thus inducing oxidative stress and impaired insulin signaling in 3T3-L1 adipocytes. This suggests that TNFα treatment can be used as a basic in vitro model for studying the pathophysiology of mitochondrial dysfunction and related metabolic complications and screening potential anti-obesity therapeutics in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Babalwa Unice Jack
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa.
- Centre for Cardiometabolic Research in Africa, Division of Medical Physiology, Stellenbosch University, Tygerberg, Cape Town, 7505, South Africa.
| | - Stephanie Dias
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa
- Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of Pretoria, Pretoria, 0001, South Africa
| |
Collapse
|
8
|
Rudolph EL, Chin L. Mechanobiology in Metabolic Dysfunction-Associated Steatotic Liver Disease and Obesity. Curr Issues Mol Biol 2024; 46:7134-7146. [PMID: 39057066 PMCID: PMC11276231 DOI: 10.3390/cimb46070425] [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: 06/01/2024] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
With the ongoing obesity epidemic, the prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is expected to rise and necessitates a greater understanding of how the disease proceeds from benign excess lipid in hepatocytes to liver fibrosis and eventually to liver cancer. MASLD is caused, at least in part, by hepatocytes' storage of free fatty acids (FAs) that dysfunctional adipocytes are no longer able to store, and therefore, MASLD is a disease that involves both the liver and adipose tissues. The disease progression is not only facilitated by biochemical signals, but also by mechanical cues such as the increase in stiffness often seen with fibrotic fatty livers. The change in stiffness and accumulation of excess lipid droplets impact the ability of a cell to mechanosense and mechanotranduce, which perpetuates the disease. A mechanosensitive protein that is largely unexplored and could serve as a potential therapeutic target is the intermediate filament vimentin. In this review, we briefly summarize the recent research on hepatocyte and adipocyte mechanobiology and provide a synopsis of studies on the varied, and sometimes contradictory, roles of vimentin. This review is intended to benefit and encourage future studies on hepatocyte and adipocyte mechanobiology in the context of MASLD and obesity.
Collapse
Affiliation(s)
| | - LiKang Chin
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA;
| |
Collapse
|
9
|
Wang G, Muñoz-Rojas AR, Spallanzani RG, Franklin RA, Benoist C, Mathis D. Adipose-tissue Treg cells restrain differentiation of stromal adipocyte precursors to promote insulin sensitivity and metabolic homeostasis. Immunity 2024; 57:1345-1359.e5. [PMID: 38692280 PMCID: PMC11188921 DOI: 10.1016/j.immuni.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 02/08/2024] [Accepted: 04/04/2024] [Indexed: 05/03/2024]
Abstract
Regulatory T (Treg) cells in epidydimal visceral adipose tissue (eVAT) of lean mice and humans regulate metabolic homeostasis. We found that constitutive or punctual depletion of eVAT-Treg cells reined in the differentiation of stromal adipocyte precursors. Co-culture of these precursors with conditional medium from eVAT-Treg cells limited their differentiation in vitro, suggesting a direct effect. Transcriptional comparison of adipocyte precursors, matured in the presence or absence of the eVAT-Treg-conditioned medium, identified the oncostatin-M (OSM) signaling pathway as a key distinction. Addition of OSM to in vitro cultures blocked the differentiation of adipocyte precursors, while co-addition of anti-OSM antibodies reversed the ability of the eVAT-Treg-conditioned medium to inhibit in vitro adipogenesis. Genetic depletion of OSM (specifically in Treg) cells or of the OSM receptor (specifically on stromal cells) strongly impaired insulin sensitivity and related metabolic indices. Thus, Treg-cell-mediated control of local progenitor cells maintains adipose tissue and metabolic homeostasis, a regulatory axis seemingly conserved in humans.
Collapse
Affiliation(s)
- Gang Wang
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | | | | | - Ruth A Franklin
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | | | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Yang Z, Chen F, Zhang Y, Ou M, Tan P, Xu X, Li Q, Zhou S. Therapeutic targeting of white adipose tissue metabolic dysfunction in obesity: mechanisms and opportunities. MedComm (Beijing) 2024; 5:e560. [PMID: 38812572 PMCID: PMC11134193 DOI: 10.1002/mco2.560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 04/09/2024] [Accepted: 04/14/2024] [Indexed: 05/31/2024] Open
Abstract
White adipose tissue is not only a highly heterogeneous organ containing various cells, such as adipocytes, adipose stem and progenitor cells, and immune cells, but also an endocrine organ that is highly important for regulating metabolic and immune homeostasis. In individuals with obesity, dynamic cellular changes in adipose tissue result in phenotypic switching and adipose tissue dysfunction, including pathological expansion, WAT fibrosis, immune cell infiltration, endoplasmic reticulum stress, and ectopic lipid accumulation, ultimately leading to chronic low-grade inflammation and insulin resistance. Recently, many distinct subpopulations of adipose tissue have been identified, providing new insights into the potential mechanisms of adipose dysfunction in individuals with obesity. Therefore, targeting white adipose tissue as a therapeutic agent for treating obesity and obesity-related metabolic diseases is of great scientific interest. Here, we provide an overview of white adipose tissue remodeling in individuals with obesity including cellular changes and discuss the underlying regulatory mechanisms of white adipose tissue metabolic dysfunction. Currently, various studies have uncovered promising targets and strategies for obesity treatment. We also outline the potential therapeutic signaling pathways of targeting adipose tissue and summarize existing therapeutic strategies for antiobesity treatment including pharmacological approaches, lifestyle interventions, and novel therapies.
Collapse
Affiliation(s)
- Zi‐Han Yang
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang‐Zhou Chen
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi‐Xiang Zhang
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Min‐Yi Ou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Poh‐Ching Tan
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue‐Wen Xu
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Qing‐Feng Li
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuang‐Bai Zhou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
11
|
Chen H, Luo S, Deng X, Li S, Mao Y, Yan J, Cheng Y, Liu X, Pan J, Huang H. Pre-eclamptic foetal programming predisposes offspring to hepatic steatosis via DNA methylation. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167189. [PMID: 38648899 DOI: 10.1016/j.bbadis.2024.167189] [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: 11/23/2023] [Revised: 04/01/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
OBJECTIVES Gamete and embryo-foetal origins of adult diseases hypothesis proposes that adulthood chronic disorders are associated with adverse foetal and early life traits. Our study aimed to characterise developmental changes and underlying mechanisms of metabolic disorders in offspring of pre-eclampsia (PE) programmed pregnancy. METHODS Nω-Nitro-l-arginine methyl ester hydrochloride (L-NAME) induced pre-eclampsia-like C57BL/6J mouse model was used. Lipid profiling, histological morphology, indirect calorimetry, mRNA sequencing, and pyrosequencing were performed on PE offspring of both young and elderly ages. RESULTS PE offspring exhibited increased postnatal weight gain, hepatic lipid accumulation, enlarged adipocytes, and impaired energy balance that continued to adulthood. Integrated RNA sequencing of foetal and 52-week-old livers revealed that the differentially expressed genes were mainly enriched in lipid metabolism, including glycerol-3-phosphate acyl-transferase 3 (Gpat3), a key enzyme for de novo synthesis of triglycerides (TG), and carnitine palmitoyltransferase-1a (Cpt1a), a key transmembrane enzyme that mediates fatty acid degradation. Pyrosequencing of livers from PE offspring identified hypomethylated and hypermethylated regions in Gpat3 and Cpt1a promoters, which were associated with upregulated and downregulated expressions of Gpat3 and Cpt1a, respectively. These epigenetic alterations are persistent and consistent from the foetal stage to adulthood in PE offspring. CONCLUSION These findings suggest a methylation-mediated epigenetic mechanism for PE-induced intergenerational lipid accumulation, impaired energy balance and obesity in offspring, and indicate the potential benefits of early interventions in offspring exposed to maternal PE to reduce their susceptibility to metabolic disorder in their later life.
Collapse
Affiliation(s)
- Huixi Chen
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200030, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; State Key Laboratory of Cardiology, Shanghai 200000, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Sisi Luo
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Xiuyu Deng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200000, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Sisi Li
- Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Reproductive Medicine Center, International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China
| | - Yiting Mao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Jing Yan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Yi Cheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Xia Liu
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Jiexue Pan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200030, China.
| | - Hefeng Huang
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China; Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Reproductive Medicine Center, International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200030, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; State Key Laboratory of Cardiology, Shanghai 200000, China.
| |
Collapse
|
12
|
Hong L, Zahradka P, Taylor CG. Differential Modulation by Eicosapentaenoic Acid (EPA) and Docosahexaenoic Acid (DHA) of Mesenteric Fat and Macrophages and T Cells in Adipose Tissue of Obese fa/ fa Zucker Rats. Nutrients 2024; 16:1311. [PMID: 38732558 PMCID: PMC11085824 DOI: 10.3390/nu16091311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Polyunsaturated fatty acids (PUFAs) can alter adipose tissue function; however, the relative effects of plant and marine n3-PUFAs are less clear. Our objective was to directly compare the n3-PUFAs, plant-based α-linolenic acid (ALA) in flaxseed oil, and marine-based eicosapentaenoic acid (EPA) or docosahexaenoic acid (DHA) in high-purity oils versus n6-PUFA containing linoleic acid (LA) for their effects on the adipose tissue and oral glucose tolerance of obese rats. Male fa/fa Zucker rats were assigned to faALA, faEPA, faDHA, and faLA groups and compared to baseline fa/fa rats (faBASE) and lean Zucker rats (lnLA). After 8 weeks, faEPA and faDHA had 11-14% lower body weight than faLA. The oral glucose tolerance and total body fat were unchanged, but faEPA had less mesenteric fat. faEPA and faDHA had fewer large adipocytes compared to faLA and faALA. EPA reduced macrophages in the adipose tissue of fa/fa rats compared to ALA and DHA, while faLA had the greatest macrophage infiltration. DHA decreased (~10-fold) T-cell infiltration compared to faBASE and faEPA, whereas faALA and faLA had an ~40% increase. The n3-PUFA diets attenuated tumour necrosis factor-α in adipose tissue compared to faBASE, while it was increased by LA in both genotypes. In conclusion, EPA and DHA target different aspects of inflammation in adipose tissue.
Collapse
Affiliation(s)
- Lena Hong
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada;
| | - Peter Zahradka
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada;
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Carla G. Taylor
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada;
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
13
|
Zhang Q, Lu C, Lu F, Liao Y, Cai J, Gao J. Challenges and opportunities in obesity: the role of adipocytes during tissue fibrosis. Front Endocrinol (Lausanne) 2024; 15:1365156. [PMID: 38686209 PMCID: PMC11056552 DOI: 10.3389/fendo.2024.1365156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Obesity is a chronic disease that affects the energy balance of the whole body. In addition to increasing fat mass, tissue fibrosis occurred in white adipose tissue in obese condition. Fibrosis is the over-activation of fibroblasts leading to excessive accumulation of extracellular matrix, which could be caused by various factors, including the status of adipocytes. The morphology of adipocytes responds rapidly and dynamically to nutrient fluctuations. Adaptive hypertrophy of normal adipocytes protects peripheral organs from damage from lipotoxicity. However, the biological behavior of hypertrophic adipocytes in chronic obesity is abnormally altered. Adipocytes lead to fibrotic remodeling of the extracellular matrix by inducing unresolved chronic inflammation, persistent hypoxia, and increasing myofibroblast numbers. Moreover, adipocyte-induced fibrosis not only restricts the flexible expansion and contraction of adipose tissue but also initiates the development of various diseases through cellular autonomic and paracrine effects. Regarding anti-fibrotic therapy, dysregulated intracellular signaling and epigenetic changes represent potential candidate targets. Thus, modulation of adipocytes may provide potential therapeutic avenues for reversing pathological fibrosis in adipose tissue and achieving the anti-obesity purpose.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chongxuan Lu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junrong Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Hagberg CE, Spalding KL. White adipocyte dysfunction and obesity-associated pathologies in humans. Nat Rev Mol Cell Biol 2024; 25:270-289. [PMID: 38086922 DOI: 10.1038/s41580-023-00680-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 02/10/2024]
Abstract
The prevalence of obesity and associated chronic diseases continues to increase worldwide, negatively impacting on societies and economies. Whereas the association between excess body weight and increased risk for developing a multitude of diseases is well established, the initiating mechanisms by which weight gain impairs our metabolic health remain surprisingly contested. In order to better address the myriad of disease states associated with obesity, it is essential to understand adipose tissue dysfunction and develop strategies for reinforcing adipocyte health. In this Review we outline the diverse physiological functions and pathological roles of human white adipocytes, examining our current knowledge of why white adipocytes are vital for systemic metabolic control, yet poorly adapted to our current obesogenic environment.
Collapse
Affiliation(s)
- Carolina E Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kirsty L Spalding
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
15
|
Edwin RK, Acharya LP, Maity SK, Chakrabarti P, Tantia O, Joshi MB, Satyamoorthy K, Parsa KVL, Misra P. TGS1/PIMT knockdown reduces lipid accumulation in adipocytes, limits body weight gain and promotes insulin sensitivity in mice. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166896. [PMID: 37751782 DOI: 10.1016/j.bbadis.2023.166896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 09/28/2023]
Abstract
PRIP Interacting protein with Methyl Transferase domain (PIMT/TGS1) is an integral upstream coactivator in the peroxisome proliferator-activated receptor gamma (PPARγ) transcriptional apparatus. PPARγ activation alleviates insulin resistance but promotes weight gain. Herein, we show how PIMT regulates body weight while promoting insulin sensitivity in diet induced obese mice. In vitro, we observed enhanced PIMT levels during adipogenesis. Knockdown of PIMT in 3T3-L1 results in reduced lipid accumulation and alters PPARγ regulated gene expression. Intraperitoneal injection of shPIMT lentivirus in high fat diet (HFD)-fed mice caused reduced adipose tissue size and decreased expression of lipid markers. This was accompanied by significantly lower levels of inflammation, hypertrophy and hyperplasia in the different adipose depots (eWAT and iWAT). Notably, PIMT depletion limits body weight gain in HFD-fed mice along with improved impaired oral glucose clearance. It also enhanced insulin sensitivity revealed by assessment of important insulin resistance markers and increased adiponectin levels. In addition, reduced PIMT levels did not alter the serum free fatty acid and TNFα levels. Finally, the relevance of our studies to human obesity is suggested by our finding that PIMT was upregulated in adipose tissue of obese patients along with crucial fat marker genes. We speculate that PIMT may be a potential target in maintaining energy metabolism, thus regulating obesity.
Collapse
Affiliation(s)
- Rebecca Kristina Edwin
- Centre for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad 500046, India; Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India
| | - Lavanya Prakash Acharya
- Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India
| | - Sujay K Maity
- Indian Institute of Chemical Biology (CSIR-IICB), 4, Raja Subodh Chandra Mallick Rd, Poddar Nagar, Jadavpur, Kolkata, West Bengal 700032, India
| | - Partha Chakrabarti
- Indian Institute of Chemical Biology (CSIR-IICB), 4, Raja Subodh Chandra Mallick Rd, Poddar Nagar, Jadavpur, Kolkata, West Bengal 700032, India
| | - Om Tantia
- Institute of Laparoscopic Surgery Group of Hospitals, DD - 6, Sector I, Salt Lake City, Kolkata 700064, West Bengal, India
| | - Manjunath B Joshi
- Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India
| | - Kapaettu Satyamoorthy
- Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India; SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara (SDM) University, Manjushree Nagar, Sattur, Dharwad, Karnataka 580009, India.
| | - Kishore V L Parsa
- Centre for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad 500046, India.
| | - Parimal Misra
- Centre for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad 500046, India.
| |
Collapse
|
16
|
Farhana S, Kai YC, Kadir R, Sulaiman WAW, Nordin NA, Nasir NAM. The fate of adipose tissue and adipose-derived stem cells in allograft. Cell Tissue Res 2023; 394:269-292. [PMID: 37624425 DOI: 10.1007/s00441-023-03827-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023]
Abstract
Utilizing adipose tissue and adipose-derived stem cells (ADSCs) turned into a promising field of allograft in recent years. The therapeutic potential of adipose tissue and ADSCs is governed by their molecular secretions, ability to sustain multi-differentiation and self-renewal which are pivotal in reconstructive, genetic diseases, and cosmetic goals. However, revisiting the existing functional capacity of adipose tissue and ADSCs and their intricate relationship with allograft is crucial to figure out the remarkable question of safety to use in allograft due to the growing evidence of interactions between tumor microenvironment and ADSCs. For instance, the molecular secretions of adipose tissue and ADSCs induce angiogenesis, create growth factors, and control the inflammatory response; it has now been well determined. Though the existing preclinical allograft studies gave positive feedback, ADSCs and adipose tissue are attracted by some factors of tumor stroma. Moreover, allorecognition is pivotal to allograft rejection which is carried out by costimulation in a complement-dependent way and leads to the destruction of the donor cells. However, extensive preclinical trials of adipose tissue and ADSCs in allograft at molecular level are still limited. Hence, comprehensive immunomodulatory analysis could ensure the successful allograft of adipose tissue and ADSCs avoiding the oncological risk.
Collapse
Affiliation(s)
- Sadia Farhana
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Yew Chun Kai
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Ramlah Kadir
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Wan Azman Wan Sulaiman
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Nor Asyikin Nordin
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Nur Azida Mohd Nasir
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia.
| |
Collapse
|
17
|
Kim SH, Yoon HJ, Kwon YH. Increased cholesterol uptake is associated with the altered gene expression in white adipose tissue of ApoE -/- mice fed a high-fat high-cholesterol diet. Biochem Biophys Res Commun 2023; 676:109-114. [PMID: 37506471 DOI: 10.1016/j.bbrc.2023.07.043] [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: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
Apolipoprotein E knock out (ApoE-/-) mice, the widely used model for atherosclerosis, exhibits anti-obesity characteristics due to the impaired lipoprotein internalization. Since excessive accumulation of triglycerides and cholesterol in white adipose tissue (WAT) is shown to increase the risk of metabolic diseases, we investigated the effects of dietary high-fat high-cholesterol (HFHC) on gene expression profile and the possible role of cholesterol accumulation in WAT of ApoE-/- mice. Control (CON) and HFHC diets were provided to wild-type mice (WC, WH) and ApoE-/- mice (EC, EH) for 10 weeks. Although body and WAT weights were lower in the ApoE-/- group compared to the wild-type group, increases in cholesterol and lipid peroxides in WAT were only observed in the ApoE-/- group. Transcriptome analysis revealed 3660 and 839 differentially expressed genes (DEGs) in the EC/WC and EH/WH comparison, respectively. "Thermogenesis" and "Oxidative phosphorylation" KEGG pathways were found in the EC/WC comparison, but not in the EH/WH comparison. We identified 142 and 2585 DEGs in the WH/WC and EH/EC comparison respectively, indicating a stronger effect of HFHC on WAT of ApoE-/- mice. Gene ontology analysis of DEGs revealed the association of DEGs with "Regulation of inflammatory response" term, in the EH/EC comparison, but not in the WH/WC comparison. Especially, genes encoding scavenger receptors and toll-like receptors were associated with cholesterol and lipid peroxide levels in WAT of ApoE-/- mice, but not in wild-type mice. In conclusion, changes in gene expression profile of WAT were more pronounced in ApoE-/- mice compared to wild-type mice in response to HFHC, and these altered genes were related to inflammatory response. These data suggest that increased cholesterol accumulation in WAT by dietary HFHC may play a pivotal role in the regulation of gene expression in ApoE-/- mice.
Collapse
Affiliation(s)
- Su Hyeon Kim
- Department of Food and Nutrition, Seoul National University, South Korea
| | - Hyun Jeong Yoon
- Department of Food and Nutrition, Seoul National University, South Korea
| | - Young Hye Kwon
- Department of Food and Nutrition, Seoul National University, South Korea; Research Institute of Human Ecology, Seoul National University, South Korea.
| |
Collapse
|
18
|
Bentanachs R, Blanco L, Montesinos M, Sala-Vila A, Lázaro I, Rodríguez-Morató J, Sánchez RM, Laguna JC, Roglans N, Alegret M. Adipose Tissue Protects against Hepatic Steatosis in Male Rats Fed a High-Fat Diet plus Liquid Fructose: Sex-Related Differences. Nutrients 2023; 15:3909. [PMID: 37764693 PMCID: PMC10534325 DOI: 10.3390/nu15183909] [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: 07/26/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic fatty liver disease is a sexual dimorphic disease, with adipose tissue playing an essential role. Our previous work showed that female rats fed a high-fat high-fructose diet devoid of cholesterol (HFHFr) developed simple hepatic steatosis dissociated from obesity. This study assessed the impact of the HFHFr diet on the male rat metabolism compared with data obtained for female rats. A total of 16 Sprague Dawley (SD) male rats were fed either a control (standard rodent chow and water) or HFHFr (high-fat diet devoid of cholesterol, plus 10% fructose in drinking water) diet for 3 months. Unlike female rats, and despite similar increases in energy consumption, HFHFr males showed increased adiposity and hyperleptinemia. The expression of hormone-sensitive lipase in the subcutaneous white adipose tissue was enhanced, leading to high free fatty acid and glycerol serum levels. HFHFr males presented hypertriglyceridemia, but not hepatic steatosis, partially due to enhanced liver PPARα-related fatty acid β-oxidation and the VLDL-promoting effect of leptin. In conclusion, the SD rats showed a sex-related dimorphic response to the HFHFr diet. Contrary to previous results for HFHFr female rats, the male rats were able to expand the adipose tissue, increase fatty acid catabolism, or export it as VLDL, avoiding liver lipid deposition.
Collapse
Affiliation(s)
- Roger Bentanachs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (R.B.); (L.B.); (M.M.); (R.M.S.); (J.C.L.)
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Laia Blanco
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (R.B.); (L.B.); (M.M.); (R.M.S.); (J.C.L.)
| | - Maria Montesinos
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (R.B.); (L.B.); (M.M.); (R.M.S.); (J.C.L.)
| | - Aleix Sala-Vila
- IMIM-Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain; (A.S.-V.); (I.L.)
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Iolanda Lázaro
- IMIM-Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain; (A.S.-V.); (I.L.)
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Jose Rodríguez-Morató
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Rosa María Sánchez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (R.B.); (L.B.); (M.M.); (R.M.S.); (J.C.L.)
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Juan Carlos Laguna
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (R.B.); (L.B.); (M.M.); (R.M.S.); (J.C.L.)
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Núria Roglans
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (R.B.); (L.B.); (M.M.); (R.M.S.); (J.C.L.)
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Marta Alegret
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (R.B.); (L.B.); (M.M.); (R.M.S.); (J.C.L.)
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| |
Collapse
|
19
|
Kruppa P, Gohlke S, Łapiński K, Garcia-Carrizo F, Soultoukis GA, Infanger M, Schulz TJ, Ghods M. Lipedema stage affects adipocyte hypertrophy, subcutaneous adipose tissue inflammation and interstitial fibrosis. Front Immunol 2023; 14:1223264. [PMID: 37575263 PMCID: PMC10417720 DOI: 10.3389/fimmu.2023.1223264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Lipedema is a painful subcutaneous adipose tissue (SAT) disease characterized by adipocyte hypertrophy, immune cell recruitment, and fibrosis in the affected areas. These features are thought to contribute to the development and progression of the condition. However, the relationship between lipedema disease stage and the associated adipose tissue changes has not been determined so far. Methods SAT biopsies of 32 lipedema patients, ranging across the pathological stages I to III, and 14 BMI- and age-matched controls were harvested from lipedema-affected thighs and non-symptomatic lower abdominal regions. Histological and immunohistochemical (IHC) staining and expression analysis of markers for adipogenesis, immunomodulation, and fibrosis were performed on the tissue biopsies. Results Lipedema patients showed increased adipocyte areas and a stage-dependent shift towards larger cell sizes in the thighs. Lipedema SAT was linked with increased interstitial collagen accumulation in the thighs, but not the lower abdominal region when compared to controls. There was a trend toward progressive SAT fibrosis of the affected thighs with increasing lipedema stage. Elevated gene expression levels of macrophage markers were found for thigh SAT biopsies, but not in the abdominal region. IHC staining of lipedema thigh biopsies confirmed a transiently elevated macrophage polarization towards an M2-like (anti-inflammatory) phenotype. Conclusions In summary, lipedema SAT is associated with stage-dependent adipocyte hypertrophy, stage-progressive interstitial fibrosis and elevated proportion of M2-like macrophages. The character of the inflammatory response differs from primary obesity and may possess an essential role in the development of lipedema.
Collapse
Affiliation(s)
- Philipp Kruppa
- Department of Plastic, Aesthetic and Reconstructive Microsurgery/Hand Surgery, Hospital Ernst von Bergmann, Potsdam, Germany
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition (DIfE) Potsdam-Rehbrücke, Nuthetal, Germany
- Otto-von-Guericke University Magdeburg, Department of Plastic, Aesthetic and Hand Surgery, Magdeburg, Germany
| | - Sabrina Gohlke
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition (DIfE) Potsdam-Rehbrücke, Nuthetal, Germany
| | - Kamila Łapiński
- Department of Plastic, Aesthetic and Reconstructive Microsurgery/Hand Surgery, Hospital Ernst von Bergmann, Potsdam, Germany
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition (DIfE) Potsdam-Rehbrücke, Nuthetal, Germany
- Otto-von-Guericke University Magdeburg, Department of Plastic, Aesthetic and Hand Surgery, Magdeburg, Germany
| | - Francisco Garcia-Carrizo
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition (DIfE) Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München, Germany
| | - George A. Soultoukis
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition (DIfE) Potsdam-Rehbrücke, Nuthetal, Germany
| | - Manfred Infanger
- Otto-von-Guericke University Magdeburg, Department of Plastic, Aesthetic and Hand Surgery, Magdeburg, Germany
| | - Tim J. Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition (DIfE) Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Mojtaba Ghods
- Department of Plastic, Aesthetic and Reconstructive Microsurgery/Hand Surgery, Hospital Ernst von Bergmann, Potsdam, Germany
| |
Collapse
|
20
|
Bellot PENR, Braga ES, Omage FB, da Silva Nunes FL, Lima SCVC, Lyra CO, Marchioni DML, Pedrosa LFC, Barbosa F, Tasic L, Sena-Evangelista KCM. Plasma lipid metabolites as potential biomarkers for identifying individuals at risk of obesity-induced metabolic complications. Sci Rep 2023; 13:11729. [PMID: 37474543 PMCID: PMC10359283 DOI: 10.1038/s41598-023-38703-8] [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: 12/28/2022] [Accepted: 07/13/2023] [Indexed: 07/22/2023] Open
Abstract
Lipidomics studies have indicated an association between obesity and lipid metabolism dysfunction. This study aimed to evaluate and compare cardiometabolic risk factors, and the lipidomic profile in adults and older people. A cross-sectional study was conducted with 72 individuals, divided into two sex and age-matched groups: obese (body mass index-BMI ≥ 30 kg/m2; n = 36) and non-obese (BMI < 30 kg/m2; n = 36). The lipidomic profiles were evaluated in plasma using 1H nuclear magnetic resonance (1H-NMR) spectroscopy. Obese individuals had higher waist circumference (p < 0.001), visceral adiposity index (p = 0.029), homeostatic model assessment insulin resistance (HOMA-IR) (p = 0.010), and triacylglycerols (TAG) levels (p = 0.018). 1H-NMR analysis identified higher amounts of saturated lipid metabolite fragments, lower levels of unsaturated lipids, and some phosphatidylcholine species in the obese group. Two powerful machine learning (ML) models-k-nearest neighbors (kNN) and XGBoost (XGB) were employed to characterize the lipidomic profile of obese individuals. The results revealed metabolic alterations associated with obesity in the NMR signals. The models achieved high accuracy of 86% and 81%, respectively. The feature importance analysis identified signal at 1.50-1.60 ppm (-CO-CH2-CH2-, Cholesterol and fatty acid in TAG, Phospholipids) to have the highest importance in the two models.
Collapse
Affiliation(s)
- Paula Emília Nunes Ribeiro Bellot
- Postgraduate Program in Nutrition, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Erik Sobrinho Braga
- Biological Chemistry Laboratory, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Folorunsho Bright Omage
- Biological Chemistry Laboratory, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Computational Biology Research Group, Embrapa Agricultural Informatics, Campinas, São Paulo, Brazil
| | - Francisca Leide da Silva Nunes
- Postgraduate Program in Nutrition, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | | | - Clélia Oliveira Lyra
- Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Dirce Maria Lobo Marchioni
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo Campus, São Paulo, SP, Brazil
| | | | - Fernando Barbosa
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto of the University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ljubica Tasic
- Biological Chemistry Laboratory, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | | |
Collapse
|
21
|
Lam CS, Xia YX, Chen BS, Du YX, Liu KL, Zhang HJ. Dihydro-Resveratrol Attenuates Oxidative Stress, Adipogenesis and Insulin Resistance in In Vitro Models and High-Fat Diet-Induced Mouse Model via AMPK Activation. Nutrients 2023; 15:3006. [PMID: 37447331 DOI: 10.3390/nu15133006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Management of obesity has become a prevalent strategy for preventing the diseases closely integrated with excess body weight such as diabetes over the last half century. Searching for therapeutic agents acting on oxidative stress, adipogenesis and insulin resistance is considered as an efficient approach to control obesity-related diseases. The present study was designed to examine the in vitro and in vivo effects of dihydro-resveratrol (DR2), a naturally occurring compound from Dendrobium medicinal plants, on oxidative stress aggravation, adipogenesis, lipogenesis and insulin sensitivity. We utilized an in vitro 3T3-L1 adipocyte differentiation model to show that DR2 could reduce pre-adipocyte maturation by activation of AMPK/SIRT1 signaling proteins to inhibit p38MAPK proteins. With the use of in vitro oxidative-stress-induced hepatocytes and myoblasts models, DR2 was also shown to be able to reduce oxidative stress aggravation through mediation of Nrf2-related antioxidative cascade, reduce intracellular lipid accumulation through phosphorylation of ACC protein, reduce lipid peroxidation in hepatocytes and promote insulin sensitivity via activation of AKT protein in the insulin-resistant HepG2 cells and C2C12 cells. The effects of DR2 on adipogenesis, lipid accumulation, insulin resistance and blood glucose clearance were further demonstrated in the high-fat diet-induced obesity mouse model. Our in vitro and in vivo studies determined that DR2 could contain therapeutic potential for the treatment of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Chu-Shing Lam
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Yi-Xuan Xia
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Bai-Sen Chen
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Yin-Xiao Du
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Kang-Lun Liu
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Hong-Jie Zhang
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China
| |
Collapse
|
22
|
Chen M, Dong Y, Tian L, Zhou J, Zhu E, Yuan H, Li X, Wang B. Metastasis suppressor 1 interacts with protein tyrosine phosphatase receptor-δ to regulate adipogenesis. FASEB J 2023; 37:e22857. [PMID: 36906292 DOI: 10.1096/fj.202201322r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 02/07/2023] [Accepted: 02/22/2023] [Indexed: 03/13/2023]
Abstract
Adipogenesis is a finely controlled process and its dysfunction may contribute to metabolic disorders such as obesity. Metastasis suppressor 1 (MTSS1) is a player in tumorigenesis and metastasis of various types of cancers. To date, it is not known whether and how MTSS1 plays a role in adipocyte differentiation. In the current study, we found that MTSS1 was upregulated during adipogenic differentiation of established mesenchymal cell lines and primary cultured bone marrow stromal cells. Gain-of-function and loss-of-function experiments uncovered that MTSS1 facilitated adipocyte differentiation from mesenchymal progenitor cells. Mechanistic explorations revealed that MTSS1 bound and interacted with FYN, a member of Src family of tyrosine kinases (SFKs), and protein tyrosine phosphatase receptor-δ (PTPRD). We demonstrated that PTPRD was capable of inducing the differentiation of adipocytes. Overexpression of PTPRD attenuated the impaired adipogenesis induced by the siRNA targeting MTSS1. Both MTSS1 and PTPRD activated SFKs by suppressing the phosphorylation of SFKs at Tyr530 and inducing the phosphorylation of FYN at Tyr419. Further investigation showed that MTSS1 and PTPRD were able to activate FYN. Collectively, our study has for the first time unraveled that MTSS1 plays a role in adipocyte differentiation in vitro through interacting with PTPRD and thereby activating SFKs such as FYN tyrosine kinase.
Collapse
Affiliation(s)
- Meng Chen
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Yuan Dong
- College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lijie Tian
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Jie Zhou
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Endong Zhu
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Hairui Yuan
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiaoxia Li
- College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Baoli Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
23
|
Santillana N, Astudillo-Guerrero C, D’Espessailles A, Cruz G. White Adipose Tissue Dysfunction: Pathophysiology and Emergent Measurements. Nutrients 2023; 15:nu15071722. [PMID: 37049561 PMCID: PMC10096946 DOI: 10.3390/nu15071722] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
White adipose tissue (AT) dysfunction plays an important role in the development of cardiometabolic alterations associated with obesity. AT dysfunction is characterized by the loss of the expansion capacity of the AT, an increment in adipocyte hypertrophy, and changes in the secretion profile of adipose cells, associated with accumulation of macrophages and inflammation. Since not all people with an excess of adiposity develop comorbidities, it is necessary to find simple tools that can evidence AT dysfunction and allow the detection of those people with the potential to develop metabolic alterations. This review focuses on the current pathophysiological mechanisms of white AT dysfunction and emerging measurements to assess its functionality.
Collapse
Affiliation(s)
- Natalia Santillana
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago 8380453, Chile
| | - Camila Astudillo-Guerrero
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Amanda D’Espessailles
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| |
Collapse
|
24
|
Bhardwaj P, Iyengar NM, Zahid H, Carter KM, Byun DJ, Choi MH, Sun Q, Savenkov O, Louka C, Liu C, Piloco P, Acosta M, Bareja R, Elemento O, Foronda M, Dow LE, Oshchepkova S, Giri DD, Pollak M, Zhou XK, Hopkins BD, Laughney AM, Frey MK, Ellenson LH, Morrow M, Spector JA, Cantley LC, Brown KA. Obesity promotes breast epithelium DNA damage in women carrying a germline mutation in BRCA1 or BRCA2. Sci Transl Med 2023; 15:eade1857. [PMID: 36812344 PMCID: PMC10557057 DOI: 10.1126/scitranslmed.ade1857] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 01/23/2023] [Indexed: 02/24/2023]
Abstract
Obesity, defined as a body mass index (BMI) ≥ 30, is an established risk factor for breast cancer among women in the general population after menopause. Whether elevated BMI is a risk factor for women with a germline mutation in BRCA1 or BRCA2 is less clear because of inconsistent findings from epidemiological studies and a lack of mechanistic studies in this population. Here, we show that DNA damage in normal breast epithelia of women carrying a BRCA mutation is positively correlated with BMI and with biomarkers of metabolic dysfunction. In addition, RNA sequencing showed obesity-associated alterations to the breast adipose microenvironment of BRCA mutation carriers, including activation of estrogen biosynthesis, which affected neighboring breast epithelial cells. In breast tissue explants cultured from women carrying a BRCA mutation, we found that blockade of estrogen biosynthesis or estrogen receptor activity decreased DNA damage. Additional obesity-associated factors, including leptin and insulin, increased DNA damage in human BRCA heterozygous epithelial cells, and inhibiting the signaling of these factors with a leptin-neutralizing antibody or PI3K inhibitor, respectively, decreased DNA damage. Furthermore, we show that increased adiposity was associated with mammary gland DNA damage and increased penetrance of mammary tumors in Brca1+/- mice. Overall, our results provide mechanistic evidence in support of a link between elevated BMI and breast cancer development in BRCA mutation carriers. This suggests that maintaining a lower body weight or pharmacologically targeting estrogen or metabolic dysfunction may reduce the risk of breast cancer in this population.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Neil M. Iyengar
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Heba Zahid
- Department of Medical Laboratory Technology, College of Applied Medical Science, Taibah University, Medina 42353, Saudi Arabia
| | | | - Dong Jun Byun
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Man Ho Choi
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Qi Sun
- Computational Biology Service Unit of Life Sciences Core Laboratories Center, Cornell University, Ithaca, NY 14853, USA
| | - Oleksandr Savenkov
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, USA
| | - Charalambia Louka
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Catherine Liu
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Phoebe Piloco
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Monica Acosta
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Rohan Bareja
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Miguel Foronda
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lukas E. Dow
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sofya Oshchepkova
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Dilip D. Giri
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael Pollak
- Departments of Medicine and Oncology, McGill University, Montreal, Canada
| | - Xi Kathy Zhou
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, USA
| | - Benjamin D. Hopkins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ashley M. Laughney
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Melissa K. Frey
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lora Hedrick Ellenson
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Monica Morrow
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jason A. Spector
- Laboratory of Bioregenerative Medicine and Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lewis C. Cantley
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
25
|
Zhu M, Li H, Yin Y, Ding M, Philips CA, Romeiro FG, Qi X. U-shaped relationship between subcutaneous adipose tissue index and mortality in liver cirrhosis. J Cachexia Sarcopenia Muscle 2023; 14:508-516. [PMID: 36577511 PMCID: PMC9891908 DOI: 10.1002/jcsm.13154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/03/2022] [Accepted: 11/29/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Subcutaneous and visceral adipose tissues are important body components, but their effects on the mortality in patients with liver cirrhosis remain controversial based on the current evidence. METHODS We retrospectively identified 372 eligible patients in whom subcutaneous adipose tissue index (SATI) and visceral adipose tissue index (VATI) could be measured by computed tomography images at the third lumbar vertebra. The association of SATI and VATI with the risk of death was evaluated on a continuous scale with restricted cubic spline curves based on Cox proportional hazards models. Cumulative probability of mortality was estimated by Nelson-Aalen cumulative risk curve analyses. Independent predictors of death were evaluated by competing risk analyses after adjusting for age, sex, and model for end-stage liver disease score. RESULTS Majority of patients were male (69.4%) with a mean age of 55.40 ± 10.68 years. SATI had a U-shaped association with mortality (P for non-linearity <0.001). Cutoff values of SATI were 19.7 and 51.8 cm2 /m2 at the points where hazard ratios were just <1.2. SATI was categorized as low (<19.7 cm2 /m2 ), moderate (19.7-51.8 cm2 /m2 ), and high (>51.8 cm2 /m2 ) level. There was no significant difference in the cumulative probability of mortality between low versus moderate SATI groups (Gray's test, P = 0.052) and high versus moderate SATI groups (Gray's test, P = 0.054). Competing risk analyses demonstrated that low SATI could increase the mortality compared with moderate SATI (subdistribution hazard ratio [sHR] = 1.66, 95% confidence interval [CI]: 0.992-2.78, P = 0.054) and was an independent predictor of death (sHR = 1.86, 95% CI: 1.059-3.28, P = 0.031). Competing risk analyses also demonstrated that high SATI could significantly increase the mortality compared with moderate SATI (sHR = 1.6, 95% CI: 1-2.54, P = 0.049), and was an independent predictor of death (sHR = 2.007, 95% CI: 1.195-3.37, P = 0.0085). VATI had an irregularly shaped association with mortality (P for non-linearity <0.001). Cutoff values of VATI were 9.8 and 40.2 cm2 /m2 at the points where hazard ratios were just <1.2. VATI was categorized as low (<9.8 cm2 /m2 ), moderate (9.8-40.2 cm2 /m2 ), and high (>40.2 cm2 /m2 ) level. There was no significant difference in the cumulative probability of mortality between low versus moderate VATI groups (Gray's test, P = 0.381) and high versus moderate VATI groups (Gray's test, P = 0.787). Competing risk analyses demonstrated that neither low (sHR = 1.27, 95% CI: 0.599-2.7, P = 0.53) nor high VATI (sHR = 0.848, 95% CI: 0.539-1.34, P = 0.48) was an independent predictor of death compared with moderate VATI. CONCLUSIONS Both excessive deficiency and accumulation of subcutaneous adipose tissues negatively influence the outcomes of cirrhotic patients.
Collapse
Affiliation(s)
- Menghua Zhu
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang, China.,Postgraduate College, Jinzhou Medical University, Jinzhou, China
| | - Hongyu Li
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang, China.,Postgraduate College, Jinzhou Medical University, Jinzhou, China.,Postgraduate College, China Medical University, Shenyang, China
| | - Yue Yin
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang, China.,Postgraduate College, China Medical University, Shenyang, China
| | - Min Ding
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang, China.,Postgraduate College, China Medical University, Shenyang, China
| | - Cyriac Abby Philips
- Department of Clinical and Translational Hepatology, The Liver Institute, Center of Excellence in GI Sciences, Rajagiri Hospital, Aluva, Kerala, India
| | | | - Xingshun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang, China.,Postgraduate College, Jinzhou Medical University, Jinzhou, China.,Postgraduate College, China Medical University, Shenyang, China
| |
Collapse
|
26
|
de la Monte SM. Malignant Brain Aging: The Formidable Link Between Dysregulated Signaling Through Mechanistic Target of Rapamycin Pathways and Alzheimer's Disease (Type 3 Diabetes). J Alzheimers Dis 2023; 95:1301-1337. [PMID: 37718817 PMCID: PMC10896181 DOI: 10.3233/jad-230555] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Malignant brain aging corresponds to accelerated age-related declines in brain functions eventually derailing the self-sustaining forces that govern independent vitality. Malignant brain aging establishes the path toward dementing neurodegeneration, including Alzheimer's disease (AD). The full spectrum of AD includes progressive dysfunction of neurons, oligodendrocytes, astrocytes, microglia, and the microvascular systems, and is mechanistically driven by insulin and insulin-like growth factor (IGF) deficiencies and resistances with accompanying deficits in energy balance, increased cellular stress, inflammation, and impaired perfusion, mimicking the core features of diabetes mellitus. The underlying pathophysiological derangements result in mitochondrial dysfunction, abnormal protein aggregation, increased oxidative and endoplasmic reticulum stress, aberrant autophagy, and abnormal post-translational modification of proteins, all of which are signature features of both AD and dysregulated insulin/IGF-1-mechanistic target of rapamycin (mTOR) signaling. This article connects the dots from benign to malignant aging to neurodegeneration by reviewing the salient pathologies associated with initially adaptive and later dysfunctional mTOR signaling in the brain. Effective therapeutic and preventive measures must be two-pronged and designed to 1) address complex and shifting impairments in mTOR signaling through the re-purpose of effective anti-diabetes therapeutics that target the brain, and 2) minimize the impact of extrinsic mediators of benign to malignant aging transitions, e.g., inflammatory states, obesity, systemic insulin resistance diseases, and repeated bouts of general anesthesia, by minimizing exposures or implementing neuroprotective measures.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Pathology and Laboratory Medicine, Medicine, Neurology and Neurosurgery, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
27
|
Zhang YX, Ou MY, Yang ZH, Sun Y, Li QF, Zhou SB. Adipose tissue aging is regulated by an altered immune system. Front Immunol 2023; 14:1125395. [PMID: 36875140 PMCID: PMC9981968 DOI: 10.3389/fimmu.2023.1125395] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/30/2023] [Indexed: 02/19/2023] Open
Abstract
Adipose tissue is a widely distributed organ that plays a critical role in age-related physiological dysfunctions as an important source of chronic sterile low-grade inflammation. Adipose tissue undergoes diverse changes during aging, including fat depot redistribution, brown and beige fat decrease, functional decline of adipose progenitor and stem cells, senescent cell accumulation, and immune cell dysregulation. Specifically, inflammaging is common in aged adipose tissue. Adipose tissue inflammaging reduces adipose plasticity and pathologically contributes to adipocyte hypertrophy, fibrosis, and ultimately, adipose tissue dysfunction. Adipose tissue inflammaging also contributes to age-related diseases, such as diabetes, cardiovascular disease and cancer. There is an increased infiltration of immune cells into adipose tissue, and these infiltrating immune cells secrete proinflammatory cytokines and chemokines. Several important molecular and signaling pathways mediate the process, including JAK/STAT, NFκB and JNK, etc. The roles of immune cells in aging adipose tissue are complex, and the underlying mechanisms remain largely unclear. In this review, we summarize the consequences and causes of inflammaging in adipose tissue. We further outline the cellular/molecular mechanisms of adipose tissue inflammaging and propose potential therapeutic targets to alleviate age-related problems.
Collapse
Affiliation(s)
- Yi-Xiang Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min-Yi Ou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zi-Han Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Sun
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qing-Feng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuang-Bai Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Davies DM, van den Handel K, Bharadwaj S, Lengefeld J. Cellular enlargement - A new hallmark of aging? Front Cell Dev Biol 2022; 10:1036602. [PMID: 36438561 PMCID: PMC9688412 DOI: 10.3389/fcell.2022.1036602] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2023] Open
Abstract
Years of important research has revealed that cells heavily invest in regulating their size. Nevertheless, it has remained unclear why accurate size control is so important. Our recent study using hematopoietic stem cells (HSCs) in vivo indicates that cellular enlargement is causally associated with aging. Here, we present an overview of these findings and their implications. Furthermore, we performed a broad literature analysis to evaluate the potential of cellular enlargement as a new aging hallmark and to examine its connection to previously described aging hallmarks. Finally, we highlight interesting work presenting a correlation between cell size and age-related diseases. Taken together, we found mounting evidence linking cellular enlargement to aging and age-related diseases. Therefore, we encourage researchers from seemingly unrelated areas to take a fresh look at their data from the perspective of cell size.
Collapse
Affiliation(s)
- Daniel M. Davies
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kim van den Handel
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Soham Bharadwaj
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jette Lengefeld
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
29
|
Muñoz-Manrique C, Trejo-Valdivia B, Hernández-Cordero S, Cantoral A, Deierlein AL, Colicino E, Niedzwiecki MM, Wright RO, Baccarelli AA, Téllez-Rojo MM. Weight gain trajectories patterns from pregnancy to early postpartum: identifying women at risk and timing to prevent weight regain. BMC Pregnancy Childbirth 2022; 22:811. [PMID: 36333677 PMCID: PMC9635073 DOI: 10.1186/s12884-022-05154-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Woman's weight changes during pregnancy and postpartum contribute to obesity and health outcomes later in life. This study aimed to identify and characterize weight change trajectories from pregnancy to one year postpartum among adult women. METHODS We used data from an ongoing cohort of healthy adult women (n = 819) with singleton pregnancies from 2007 - 2011. Sociodemographic data, pre-pregnancy body weight, and sedentary and breastfeeding practices were collected using questionaries applied by trained professionals. We applied a group-based trajectory modeling to distinguish weight change measured in the second and third trimesters of pregnancy and at one month, six, and 12 months postpartum. Multinomial regression models were run to characterize each trajectory. RESULTS We identified six weight change trajectories with the main difference in the patterns followed after one month of delivery. One in three women (36.7%) was classified in some of the three postpartum weight gain trajectories and regained weight from the second trimester of the first year postpartum. Women who followed some of these trajectories were more likely to have higher age, obesity before pregnancy, < 10 years of schooling, and partner, compared with women (10.7%, n = 87) in a postpartum sustained-fast-lost-weight trajectory (p < 0.05). CONCLUSIONS Women with obesity before pregnancy have higher odds of regaining gestational weight after delivery without reaching their pre-pregnancy weight. The first six months postpartum are crucial to establishing obesity prevention strategies. Further research is needed to evaluate the effect of the interventions that prevent substantial weight gain through reproductive years in high-risk women.
Collapse
Affiliation(s)
- Cinthya Muñoz-Manrique
- Department of Nutrition and Bioprogramming, National Institute of Perinatology, Mexico City, Mexico
| | - Belem Trejo-Valdivia
- Centro de Investigación en Nutrición y Salud, Instituto Nacional de Salud Pública, 3226, Avenida Universidad 655, Santa María Ahuacatitlán, Morelos, 62100, Cuernavaca, México.
| | - Sonia Hernández-Cordero
- Research Center for Equitable Development EQUIDE, Universidad Iberoamericana, Ciudad de México, México
| | | | - Andrea L Deierlein
- Department of Epidemiology, School of Global Public Health, New York University, New York, NY, USA
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Megan M Niedzwiecki
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Science, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | - Martha María Téllez-Rojo
- Centro de Investigación en Nutrición y Salud, Instituto Nacional de Salud Pública, 3226, Avenida Universidad 655, Santa María Ahuacatitlán, Morelos, 62100, Cuernavaca, México
| |
Collapse
|
30
|
Hong J, Kim Y. Fatty Liver/Adipose Tissue Dual-Targeting Nanoparticles with Heme Oxygenase-1 Inducer for Amelioration of Obesity, Obesity-Induced Type 2 Diabetes, and Steatohepatitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203286. [PMID: 36209391 PMCID: PMC9685446 DOI: 10.1002/advs.202203286] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/15/2022] [Indexed: 05/28/2023]
Abstract
Persistent uptake of high-calorie diets induces the storage of excessive lipid in visceral adipose tissue. Lipids secreted from obese adipose tissue are accumulated in peripheral tissues such as the liver, pancreas, and muscle, and impair insulin sensitivity causing type 2 diabetes mellitus (T2DM). Furthermore, the accumulation of inflammatory cytokines and lipids in the liver induces apoptosis and fibrogenesis, and ultimately causes nonalcoholic steatohepatitis (NASH). To modulate obese tissue environments, it is challenged to selectively deliver inducers of heme oxygenase-1 (HO-1) to adipose tissue with the aid of a prohibitin targeting drug delivery system. Prohibitin binding peptide (PBP), an oligopeptide targeting prohibitin rich in adipose tissue, is conjugated on the surface of Hemin- or CoPP-loaded poly(lactide-co-glycolide) nanoparticles (PBP-NPs). PBP-NPs efficiently differentiate lipid storing white adipocytes into energy-generating brown adipocytes in T2DM and NASH models. In addition, PBP-NPs are found to target prohibitin overexpressed fatty liver in the NASH model and inhibit hepatic uptake of circulating lipids. Furthermore, PBP-NPs switch phenotypes of inflammatory macrophages in damaged organs and lower inflammation. Taken together, dual-targeted induction of HO-1 in fatty adipose and liver tissues is proven to be a promising therapeutic strategy to ameliorate obesity, insulin resistance, and steatohepatitis by lowering lipids and cytokines.
Collapse
Affiliation(s)
- Juhyeong Hong
- Department of BioengineeringInstitute for Bioengineering and Biopharmaceutical Research Hanyang UniversitySeoul04763South Korea
- Education and Research Group for Biopharmaceutical Innovation LeaderHanyang UniversitySeoul04763South Korea
| | - Yong‐Hee Kim
- Department of BioengineeringInstitute for Bioengineering and Biopharmaceutical Research Hanyang UniversitySeoul04763South Korea
- Education and Research Group for Biopharmaceutical Innovation LeaderHanyang UniversitySeoul04763South Korea
| |
Collapse
|
31
|
Lim G, Lim Y. Effects of Whey Peptide Supplementation on Sarcopenic Obesity in High-Fat Diet-Fed Mice. Nutrients 2022; 14:4402. [PMID: 36297085 PMCID: PMC9611493 DOI: 10.3390/nu14204402] [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] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
The incidence of sarcopenic obesity gradually increased in parallel with the aged population. This research examined the effects of whey peptide (WP) supplementation with/without resistant exercise (RE) on sarcopenic obesity. Male 8-month-old C57BL/6J mice were fed a control diet (10 kcal% fat) or a high-fat diet (60 kcal% fat) for 8 weeks. High-fat diet-fed mice were randomly divided into four groups: obesity control group (OB), RE (RE only), WP (WP only), and WPE (RE and WP). WP supplementation (1500 mg/day/kg B.W.) gavage and RE (ladder climbing, five times weekly, 8−10 repetitions, 10−20% B.W. load) were conducted for an additional 8 weeks. Protein and mRNA levels of markers related to energy, protein, and lipid metabolism were analyzed in skeletal muscle and adipose tissue by one-way analysis of variance (ANOVA). WP supplementation regardless of RE significantly suppressed the increasing fat mass (p = 0.016) and decreasing lean mass (p = 0.014) and alleviated abnormal morphological changes in skeletal muscle and adipose tissue (p < 0.001). In adipose tissue, WP supplementation regardless of RE ameliorated dysregulated energy metabolism and contributed to the reduction in adipocyte differentiation (PPAR-γ (p = 0.017), C/EBPα (p = 0.034)). In skeletal muscle, WP supplementation regardless of RE alleviated energy metabolism dysregulation and resulted in down-regulated protein degradation (Atrogin-1 (p = 0.003), MuRF1 (p = 0.006)) and apoptosis (Bax) (p = 0.004). Taken together, the current study elucidated that WP supplementation regardless of RE has potential anti-obesity and anti-sarcopenic effects in sarcopenic obesity.
Collapse
Affiliation(s)
| | - Yunsook Lim
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
32
|
Men X, Han X, Lee SJ, Oh G, Park KT, Han JK, Choi SI, Lee OH. Anti-Obesogenic Effects of Sulforaphane-Rich Broccoli (Brassica oleracea var. italica) Sprouts and Myrosinase-Rich Mustard (Sinapis alba L.) Seeds in Vitro and in Vivo. Nutrients 2022; 14:nu14183814. [PMID: 36145190 PMCID: PMC9505190 DOI: 10.3390/nu14183814] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/30/2022] Open
Abstract
Glucoraphanin (GRA), a glucosinolate particularly abundant in broccoli (Brassica oleracea var. italica) sprouts, can be converted to sulforaphane (SFN) by the enzyme myrosinase. Herein, we investigated the anti-obesogenic effects of broccoli sprout powder (BSP), mustard (Sinapis alba L.) seed powder (MSP), and sulforaphane-rich MSP-BSP mixture powder (MBP) in bisphenol A (BPA)-induced 3T3-L1 cells and obese C57BL/6J mice. In vitro experiments showed that MBP, BSP, and MSP have no cytotoxic effects. Moreover, MBP and BSP inhibited the lipid accumulation in BPA-induced 3T3-L1 cells. In BPA-induced obese mice, BSP and MBP treatment inhibited body weight gain and ameliorated dyslipidemia. Furthermore, our results showed that BSP and MBP could activate AMPK, which increases ACC phosphorylation, accompanied by the upregulation of lipolysis-associated proteins (UCP-1 and CPT-1) and downregulation of adipogenesis-related proteins (C/EBP-α, FAS, aP2, PPAR-γ, and SREBP-1c), both in vitro and in vivo. Interestingly, MBP exerted a greater anti-obesogenic effect than BSP. Taken together, these findings indicate that BSP and MBP could inhibit BPA-induced adipocyte differentiation and adipogenesis by increasing the expression of the proteins related to lipid metabolism and lipolysis, effectively treating BPA-induced obesity. Thus, BSP and MBP can be developed as effective anti-obesogenic drugs.
Collapse
Affiliation(s)
- Xiao Men
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
| | - Xionggao Han
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
| | - Se-Jeong Lee
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
| | - Geon Oh
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
| | - Keun-Tae Park
- Research and Development Center, Milae Bioresourece Co., Ltd., Seoul 05542, Korea
| | - Jong-Kwon Han
- Research and Development Center, Milae Bioresourece Co., Ltd., Seoul 05542, Korea
| | - Sun-Il Choi
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
- Correspondence: (S.-I.C.); (O.-H.L.); Tel.: +82-33-250-6454 (S.-I.C.); +82-33-250-6454 (O.-H.L.); Fax: +82-33-259-5561 (S.-I.C.); +82-33-259-5561 (O.-H.L.)
| | - Ok-Hwan Lee
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
- Correspondence: (S.-I.C.); (O.-H.L.); Tel.: +82-33-250-6454 (S.-I.C.); +82-33-250-6454 (O.-H.L.); Fax: +82-33-259-5561 (S.-I.C.); +82-33-259-5561 (O.-H.L.)
| |
Collapse
|
33
|
Ostinelli G, Laforest S, Denham SG, Gauthier MF, Drolet-Labelle V, Scott E, Hould FS, Marceau S, Homer NZM, Bégin C, Andrew R, Tchernof A. Increased Adipose Tissue Indices of Androgen Catabolism and Aromatization in Women With Metabolic Dysfunction. J Clin Endocrinol Metab 2022; 107:e3330-e3342. [PMID: 35511873 PMCID: PMC9282357 DOI: 10.1210/clinem/dgac261] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Indexed: 02/02/2023]
Abstract
CONTEXT Body fat distribution is a risk factor for obesity-associated comorbidities, and adipose tissue dysfunction plays a role in this association. In humans, there is a sex difference in body fat distribution, and steroid hormones are known to regulate several cellular processes within adipose tissue. OBJECTIVE Our aim was to investigate if intra-adipose steroid concentration and expression or activity of steroidogenic enzymes were associated with features of adipose tissue dysfunction in individuals with severe obesity. METHODS Samples from 40 bariatric candidates (31 women, 9 men) were included in the study. Visceral (VAT) and subcutaneous adipose tissue (SAT) were collected during surgery. Adipose tissue morphology was measured by a combination of histological staining and semi-automated quantification. Following extraction, intra-adipose and plasma steroid concentrations were determined by liquid chromatography electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS). Aromatase activity was estimated using product over substrate ratio, while AKR1C2 activity was measured directly by fluorogenic probe. Gene expression was measured by quantitative PCR. RESULTS VAT aromatase activity was positively associated with VAT adipocyte hypertrophy (P valueadj < 0.01) and negatively with plasma high-density lipoprotein (HDL)-cholesterol (P valueadj < 0.01), while SAT aromatase activity predicted dyslipidemia in women even after adjustment for waist circumference, age, and hormonal contraceptive use. We additionally compared women with high and low visceral adiposity index (VAI) and found that VAT excess is characterized by adipose tissue dysfunction, increased androgen catabolism mirrored by increased AKR1C2 activity, and higher aromatase expression and activity indices. CONCLUSION In women, increased androgen catabolism or aromatization is associated with visceral adiposity and adipose tissue dysfunction.
Collapse
Affiliation(s)
- Giada Ostinelli
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
- École de nutrition, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sofia Laforest
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
- École de nutrition, Université Laval, Québec City, QC G1V 0A6, Canada
- University of Strathclyde, Glasgow G1 1XQ, UK
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, University/BHF, Cardiovascular Sciences, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Scott G Denham
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, University/BHF, Cardiovascular Sciences, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Marie-Frederique Gauthier
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
| | | | - Emma Scott
- Faculté de médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Frédéric-Simon Hould
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
- Faculté de médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Simon Marceau
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
- Faculté de médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, University/BHF, Cardiovascular Sciences, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Catherine Bégin
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
- École de psychologie, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Ruth Andrew
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, University/BHF, Cardiovascular Sciences, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, EH16 4TJ, UK
- BHF/CVS, Queen’s Medical Research Institute, University of Edinburgh, EH16 4TJ, UK
| | - André Tchernof
- Correspondence: Andre Tchernof, PhD, Quebec Heart and Lung Institute, School of Nutrition, Laval University, 2725 Chemin Sainte-Foy (Y-4212), Québec, QC G1V 4G5, Canada.
| |
Collapse
|
34
|
Arner P, Andersson DP, Arner E, Rydén M, Kerr AG. Subcutaneous adipose tissue expansion mechanisms are similar in early and late onset overweight/obesity. Int J Obes (Lond) 2022; 46:1196-1203. [PMID: 35228658 PMCID: PMC9151387 DOI: 10.1038/s41366-022-01102-6] [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: 09/29/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND/OBJECTIVE The development of overweight/obesity associates with alterations in white adipose tissue (WAT) cellularity (fat cell size/number) and lipid metabolism, in particular lipolysis. If these changes differ between early/juvenile (EOO < 18 years of age) or late onset overweight/obesity (LOO) is unknown and was presently examined. SUBJECTS/METHODS We included 439 subjects with validated information on body mass index (BMI) at 18 years of age. Using this information and current BMI, subjects were divided into never overweight/obese (BMI < 25 kg/m2), EOO and LOO. Adipocyte size, number, morphology (size in relation to body fat) and lipolysis were determined in subcutaneous abdominal WAT. Body composition and WAT distribution was assessed by dual-X-ray absorptiometry. RESULTS Compared with never overweight/obese, EOO and LOO displayed larger WAT amounts in all examined depots, which in subcutaneous WAT was explained by a combination of increased size and number of fat cells in EOO and LOO. EOO had 40% larger subcutaneous fat mass than LOO (p < 0.0001). Visceral WAT mass, WAT morphology and lipolysis did not differ between EOO and LOO except for minor differences in men between the two obesity groups. On average, the increase in BMI per year was 57% higher in subjects with EOO compared to LOO (p < 0.0001). CONCLUSION Early onset overweight/obesity causes a more rapid and pronounced accumulation of subcutaneous WAT than adult onset. However, fat mass expansion measures including WAT cellularity, morphology and fat cell lipolysis do not differ in an important way suggesting that similar mechanisms of WAT growth operate in EOO and LOO.
Collapse
Affiliation(s)
- Peter Arner
- Department of Medicine (H7), Karolinska Institutet at Karolinska University Hospital Huddinge, Center for Metabolism and Endocrinology, 14186, Stockholm, Sweden.
| | - Daniel P Andersson
- Department of Medicine (H7), Karolinska Institutet at Karolinska University Hospital Huddinge, Center for Metabolism and Endocrinology, 14186, Stockholm, Sweden
| | - Erik Arner
- Department of Medicine (H7), Karolinska Institutet at Karolinska University Hospital Huddinge, Center for Metabolism and Endocrinology, 14186, Stockholm, Sweden
- GSK, Gunnels Wood Rd, Stevenage, SG1 2NY, United Kingdom
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet at Karolinska University Hospital Huddinge, Center for Metabolism and Endocrinology, 14186, Stockholm, Sweden
| | - Alastair G Kerr
- Department of Medicine (H7), Karolinska Institutet at Karolinska University Hospital Huddinge, Center for Metabolism and Endocrinology, 14186, Stockholm, Sweden.
| |
Collapse
|
35
|
Banerjee D, Patra D, Sinha A, Roy S, Pant R, Sarmah R, Dutta R, Kanta Bhagabati S, Tikoo K, Pal D, Dasgupta S. Lipid-induced monokine cyclophilin-A promotes adipose tissue dysfunction implementing insulin resistance and type 2 diabetes in zebrafish and mice models of obesity. Cell Mol Life Sci 2022; 79:282. [PMID: 35511344 PMCID: PMC11072608 DOI: 10.1007/s00018-022-04306-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 11/28/2022]
Abstract
Several studies have implicated obesity-induced macrophage-adipocyte cross-talk in adipose tissue dysfunction and insulin resistance. However, the molecular cues involved in the cross-talk of macrophage and adipocyte causing insulin resistance are currently unknown. Here, we found that a lipid-induced monokine cyclophilin-A (CyPA) significantly attenuates adipocyte functions and insulin sensitivity. Targeted inhibition of CyPA in diet-induced obese zebrafish notably reduced adipose tissue inflammation and restored adipocyte function resulting in improvement of insulin sensitivity. Silencing of macrophage CyPA or pharmacological inhibition of CyPA by TMN355 effectively restored adipocytes' functions and insulin sensitivity. Interestingly, CyPA incubation markedly increased adipocyte inflammation along with an impairment of adipogenesis, however, mutation of its cognate receptor CD147 at P309A and G310A significantly waived CyPA's effect on adipocyte inflammation and its differentiation. Mechanistically, CyPA-CD147 interaction activates NF-κB signaling which promotes adipocyte inflammation by upregulating various pro-inflammatory cytokines gene expression and attenuates adipocyte differentiation by inhibiting PPARγ and C/EBPβ expression via LZTS2-mediated downregulation of β-catenin. Moreover, inhibition of CyPA or its receptor CD147 notably restored palmitate or CyPA-induced adipose tissue dysfunctions and insulin sensitivity. All these results indicate that obesity-induced macrophage-adipocyte cross-talk involving CyPA-CD147 could be a novel target for the management of insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Dipanjan Banerjee
- Metabolic Disease Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028, Assam, India
| | - Debarun Patra
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, 140001, Punjab, India
| | - Archana Sinha
- Metabolic Disease Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028, Assam, India
| | - Soumyajit Roy
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, 140001, Punjab, India
| | - Rajat Pant
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Raktim Sarmah
- Department of Aquatic Environment Management, College of Fisheries, Assam Agricultural University, Nagaon, 782103, Assam, India
| | - Rajdeep Dutta
- Department of Aquatic Environment Management, College of Fisheries, Assam Agricultural University, Nagaon, 782103, Assam, India
| | - Sarada Kanta Bhagabati
- Department of Aquatic Environment Management, College of Fisheries, Assam Agricultural University, Nagaon, 782103, Assam, India
| | - Kulbhushan Tikoo
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Durba Pal
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, 140001, Punjab, India.
| | - Suman Dasgupta
- Metabolic Disease Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028, Assam, India.
| |
Collapse
|
36
|
Mao Y, Yiran Z, Sisi L, Huixi C, Xia L, Ting W, Guolian D, Xinmei L, Sheng J, Meng Y, Huang H. Advanced paternal age increased metabolic risks in mice offspring. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166355. [DOI: 10.1016/j.bbadis.2022.166355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/07/2022] [Accepted: 01/25/2022] [Indexed: 10/19/2022]
|
37
|
Favaretto F, Bettini S, Busetto L, Milan G, Vettor R. Adipogenic progenitors in different organs: Pathophysiological implications. Rev Endocr Metab Disord 2022; 23:71-85. [PMID: 34716543 PMCID: PMC8873140 DOI: 10.1007/s11154-021-09686-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 12/14/2022]
Abstract
In physiological conditions, the adipose organ resides in well-defined areas, where it acts providing an energy supply and as an endocrine organ involved in the control of whole-body energy metabolism. Adipose tissue adipokines connect the body's nutritional status to the regulation of energy balance. When it surrounds organs, it provides also for mechanical protection. Adipose tissue has a complex and heterogenous cellular composition that includes adipocytes, adipose tissue-derived stromal and stem cells (ASCs) which are mesenchymal stromal cells, and endothelial and immune cells, which signal to each other and to other tissues to maintain homeostasis. In obesity and in other nutrition related diseases, as well as in age-related diseases, biological and functional changes of adipose tissue give rise to several complications. Obesity triggers alterations of ASCs, impairing adipose tissue remodeling and adipose tissue function, which induces low-grade systemic inflammation, progressive insulin resistance and other metabolic disorders. Adipose tissue grows by hyperplasia recruiting new ASCs and by hypertrophy, up to its expandability limit. To overcome this limitation and to store the excess of nutrients, adipose tissue develops ectopically, involving organs such as muscle, bone marrow and the heart. The origin of ectopic adipose organ is not clearly elucidated, and a possible explanation lies in the stimulation of the adipogenic differentiation of mesenchymal precursor cells which normally differentiate toward a lineage specific for the organ in which they reside. The chronic exposition of these newly-formed adipose depots to the pathological environment, will confer to them all the phenotypic characteristics of a dysfunctional adipose tissue, perpetuating the organ alterations. Visceral fat, but also ectopic fat, either in the liver, muscle or heart, can increase the risk of developing insulin resistance, type 2 diabetes, and cardiovascular diseases. Being able to prevent and to target dysfunctional adipose tissue will avoid the progression towards the complications of obesity and other nutrition-related diseases. The aim of this review is to summarize some of the knowledge regarding the presence of adipose tissue in particular tissues (where it is not usually present), describing the composition of its adipogenic precursors, and the interactions responsible for the development of organ pathologies.
Collapse
Affiliation(s)
- Francesca Favaretto
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Silvia Bettini
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Luca Busetto
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Gabriella Milan
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Roberto Vettor
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| |
Collapse
|
38
|
Majka Z, Czamara K, Janus J, Kępczyński M, Kaczor A. Prominent hypertrophy of perivascular adipocytes due to short-term high fat diet. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166315. [PMID: 34875367 DOI: 10.1016/j.bbadis.2021.166315] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/10/2021] [Accepted: 11/23/2021] [Indexed: 12/26/2022]
Abstract
Excessive lipid accumulation is a serious problem in obesity leading to adipose tissue (AT) overgrowth, chronic inflammation, endothelial dysfunction, and elevated risk of cardiovascular complications. In this work, Raman techniques coupled with fluorescence imaging were applied to characterize the effects of short-term (2 weeks) and extended (up to 8 weeks) high-fat diet (HFD) feeding on various depots of the adipose tissue of young and mature mice. Our results proved the synergistic effect of age and HFD-induced obesity manifested by changes in the morphology of adipocytes and the chemical composition of lipids. After 2 weeks of HFD feeding of young animals, substantial hypertrophy of adipocytes but only for the periaortic adipose tissue was detected with a significant decrease in lipid unsaturation degree solely in the epididymal white adipose tissue. The periaortic AT did not altered chemically due to short-term HFD feeding, however, it changed with age and with prolonged exposure to harmful factors. For older animals only brown AT remains resistant on HFD underlying its protective role and highlighting its potential as a target in obesity therapies.
Collapse
Affiliation(s)
- Zuzanna Majka
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Krzysztof Czamara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Joanna Janus
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Mariusz Kępczyński
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Agnieszka Kaczor
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland.
| |
Collapse
|
39
|
de Frutos S, Griera M, Hatem-Vaquero M, Campillo S, Gutiérrez-Calabres E, García-Ayuso D, Pardo M, Calleros L, Rodríguez-Puyol M, Rodríguez-Puyol D. The integrin beta1 modulator Tirofiban prevents adipogenesis and obesity by the overexpression of integrin-linked kinase: a pre-clinical approach in vitro and in vivo. Cell Biosci 2022; 12:10. [PMID: 35090553 PMCID: PMC8796419 DOI: 10.1186/s13578-022-00746-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/14/2022] [Indexed: 02/04/2023] Open
Abstract
Background Obesity is caused by the enlargement of the white adipose tissue (WAT) depots, characterized by the hypertrophic enlargement of malfunctioning adipocytes within WAT which increases the storage of triglycerides (TG) in the lipid droplets (LD). Adipogenesis pathways as well as the expression and activity of some extracellular matrix receptors integrins are upregulated. Integrinβ1 (INTB1) is the main isoform involved in WAT remodeling during obesity and insulin resistance-related diseases. We recently described Integrin Linked Kinase (ILK), a scaffold protein recruited by INTB1, as an important mediator of WAT remodeling and insulin resistance. As the few approved drugs to fight obesity have brought long-term cardiovascular side effects and given that the consideration of INTB1 and/or ILK modulation as anti-obesogenic strategies remains unexplored, we aimed to evaluate the anti-obesogenic capacity of the clinically approved anticoagulant Tirofiban (TF), stated in preclinical studies as a cardiovascular protector. Methods Fully differentiated adipocytes originating from C3H10T1/2 were exposed to TF and were co-treated with specific INTB1 blockers or with siRNA-based knockdown ILK expression. Lipid-specific dyes were used to determine the TG content in LD. The genetic expression pattern of ILK, pro-inflammatory cytokines (MCP1, IL6), adipogenesis (PPARγ, Leptin), thermogenesis (UCP1), proliferation (PCNA), lipid metabolism (FASN, HSL, ATGL), and metabolite transporters (FABP4, FAT, AQP7) were detected using quantitative PCR. Cytoskeletal actin polymerization was detected by confocal microscopy. Immunoblotting was performed to detect INTB1 phosphorylation at Thr788/9 and ILK activity as phosphorylation levels of protein kinase B (AKT) in Ser473 and glycogen synthase kinase 3β (GSK3β) at Ser9. TF was intraperitoneally administered once per day to wildtype and ILK knockdown mice (cKDILK) challenged with a high-fat diet (HFD) or control diet (STD) for 2 weeks. Body and WAT weight gains were compared. The expression of ILK and other markers was determined in the visceral epididymal (epi) and inguinal subcutaneous (sc) WAT. Results TF reduced TG content and the expression of adipogenesis markers and transporters in adipocytes, while UCP-1 expression was increased and the expression of lipases, cytokines or PCNA was not affected. Mechanistically, TF rapidly increased and faded the intracellular phosphorylation of INTB1 but not AKT or GSK3β. F-actin levels were rapidly decreased, and INTB1 blockade avoided the TF effect. After 24 h, ILK expression and phosphorylation rates of AKT and GSK3β were upregulated, while ILK silencing increased TG content. INTB1 blockade and ILK silencing avoided TF effects on the TG content and the transcriptional expression of PPARγ and UCP1. In HFD-challenged mice, the systemic administration of TF for several days reduced the weight gain on WAT depots. TF reduced adipogenesis and pro-inflammatory biomarkers and increased lipolysis markers HSL and FAT in epiWAT from HFD, while increased UCP1 in scWAT. In both WATs, TF upregulated ILK expression and activity, while no changes were observed in other tissues. In HFD-fed cKDILK, the blunted ILK in epiWAT worsened weight gain and avoided the anti-obesogenic effect of in vivo TF administration. Conclusions ILK downregulation in WAT can be considered a biomarker of obesity establishment. Via an INTB1-ILK axis, TF restores malfunctioning hypertrophied WAT by changing the expression of adipocyte-related genes, increasing ILK expression and activity, and reducing TG storage. TF prevents obesity, a property to be added to its anticoagulant and cardiovascular protective advantages. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00746-1.
Collapse
|
40
|
The Crowded Uterine Horn Mouse Model for Examining Postnatal Metabolic Consequences of Intrauterine Growth Restriction vs. Macrosomia in Siblings. Metabolites 2022; 12:metabo12020102. [PMID: 35208177 PMCID: PMC8880550 DOI: 10.3390/metabo12020102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 01/27/2023] Open
Abstract
Differential placental blood flow and nutrient transport can lead to both intrauterine growth restriction (IUGR) and macrosomia. Both conditions can lead to adult obesity and other conditions clustered as metabolic syndrome. We previously showed that pregnant hemi-ovariectomized mice have a crowded uterine horn, resulting in siblings whose birth weights differ by over 100% due to differential blood flow based on uterine position. We used this crowded uterus model to compare IUGR and macrosomic male mice and also identified IUGR males with rapid (IUGR-R) and low (IUGR-L) postweaning weight gain. At week 12 IUGR-R males were heavier than IUGR-L males and did not differ from macrosomic males. Rapid growth in IUGR-R males led to glucose intolerance compared to IUGR-L males and down-regulation of adipocyte signaling pathways for fat digestion and absorption and type II diabetes. Macrosomia led to increased fat mass and altered adipocyte size distribution compared to IUGR males, and down-regulation of signaling pathways for carbohydrate and fat digestion and absorption relative to IUGR-R. Clustering analysis of gonadal fat transcriptomes indicated more similarities than differences between IUGR-R and macrosomic males compared to IUGR-L males. Our findings suggest two pathways to adult metabolic disease: macrosomia and IUGR with rapid postweaning growth rate.
Collapse
|
41
|
Sex differences in white adipose tissue expansion: emerging molecular mechanisms. Clin Sci (Lond) 2021; 135:2691-2708. [PMID: 34908104 DOI: 10.1042/cs20210086] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/15/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022]
Abstract
The escalating prevalence of individuals becoming overweight and obese is a rapidly rising global health problem, placing an enormous burden on health and economic systems worldwide. Whilst obesity has well described lifestyle drivers, there is also a significant and poorly understood component that is regulated by genetics. Furthermore, there is clear evidence for sexual dimorphism in obesity, where overall risk, degree, subtype and potential complications arising from obesity all differ between males and females. The molecular mechanisms that dictate these sex differences remain mostly uncharacterised. Many studies have demonstrated that this dimorphism is unable to be solely explained by changes in hormones and their nuclear receptors alone, and instead manifests from coordinated and highly regulated gene networks, both during development and throughout life. As we acquire more knowledge in this area from approaches such as large-scale genomic association studies, the more we appreciate the true complexity and heterogeneity of obesity. Nevertheless, over the past two decades, researchers have made enormous progress in this field, and some consistent and robust mechanisms continue to be established. In this review, we will discuss some of the proposed mechanisms underlying sexual dimorphism in obesity, and discuss some of the key regulators that influence this phenomenon.
Collapse
|
42
|
Leven AS, Gieseler RK, Schlattjan M, Schreiter T, Niedergethmann M, Baars T, Baba HA, Özçürümez MK, Sowa JP, Canbay A. Association of cell death mechanisms and fibrosis in visceral white adipose tissue with pathological alterations in the liver of morbidly obese patients with NAFLD. Adipocyte 2021; 10:558-573. [PMID: 34743657 PMCID: PMC8583086 DOI: 10.1080/21623945.2021.1982164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The role of visceral white adipose tissue (vWAT) in the progression of non-alcoholic liver disease (NAFLD) with its sub entities non-alcoholic fatty liver and steatohepatitis (NAFL; NASH) is underinvestigated. We thus explored mechanisms of fibrosis and regulated cell death in vWAT and liver tissue. In NAFLD, women displayed significantly more fibrosis in vWAT than men, and collagen 1α mRNA expression was significantly upregulated. The degrees of fibrosis in vWAT and liver tissue correlated significantly. The size of vWAT-resident adipocytes in NAFLD correlated negatively with the local degree of fibrosis. The extent of apoptosis, as measured by circulating M30, positively correlated with the degree of fibrosis in vWAT; necrosis-associated HMGB1 mRNA expression was significantly downregulated in vWAT and liver tissue; (iii) necroptosis-related RIPK-3 mRNA expression was significantly upregulated in vWAT; and autophagy-related LC3 mRNA expression was significantly downregulated in vWAT, while upregulated in the liver. Thus, the different cell death mechanisms in the vWAT in NAFLD are regulated independently while not ruling out their interaction. Fibrosis in vWAT may be associated with reduced adipocyte size and thus partially protective against NAFLD progression. Abbreviations: ATG5: autophagy related 5; BAS: bariatric surgery; BMI: body mass index; ELISA: enzyme-linked immunosorbent assay; EtOH: ethanol; FFAs: free fatty acids; HCC: hepatocellular carcinoma; HMGB1: high-mobility group box 1 protein; IHC: immunohistochemistry; IL: interleukin; LC3: microtubule-associated proteins 1A/1B light chain 3B; M30: neoepitope K18Asp396-NE displayed on the caspase-cleaved keratin 18 fragment; M65: epitope present on both caspase-cleaved and intact keratin 18; NAFL: non-alcoholic fatty liver; NAFLD: non-alcoholic fatty liver disease; NAS: NAFLD activity score; NASH: non-alcoholic steatohepatitis; NLRP3: nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain containing 3; qRT-PCR: quantitative real-time polymerase-chain reaction; r: Pearson’s correlation coefficient (r); rs: Spearman’s rank correlation coefficient; RIPK3: receptor-interacting serine/threonine-protein kinase 3; T2DM: type 2 diabetes mellitus (T2DM); TUNEL: terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling; vWAT: visceral WAT; WAT: white adipose tissue
Collapse
Affiliation(s)
- Anna-Sophia Leven
- Department of Medicine, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany.,General and Visceral Surgery, Alfried Krupp Hospital Ruettenscheid, Essen, Germany
| | - Robert K Gieseler
- Department of Medicine, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany.,Laboratory of Immunology & Molecular Biology, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany
| | - Martin Schlattjan
- Institute for Pathology, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Thomas Schreiter
- Department of Medicine, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany.,Laboratory of Immunology & Molecular Biology, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany
| | - Marco Niedergethmann
- General and Visceral Surgery, Alfried Krupp Hospital Ruettenscheid, Essen, Germany
| | - Theodor Baars
- Department of Medicine, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany.,Section of Metabolic and Preventive Medicine, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany
| | - Hideo A Baba
- Institute for Pathology, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Mustafa K Özçürümez
- Department of Medicine, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany.,Department of Laboratory Medicine, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany
| | - Jan-Peter Sowa
- Department of Medicine, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany.,Laboratory of Immunology & Molecular Biology, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany
| | - Ali Canbay
- Department of Medicine, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany.,Section of Hepatology and Gastroenterology, University Hospital, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
43
|
Jin H, Oh HJ, Nah SY, Lee BY. Gintonin-enriched fraction protects against sarcopenic obesity by promoting energy expenditure and attenuating skeletal muscle atrophy in high-fat diet-fed mice. J Ginseng Res 2021; 46:454-463. [PMID: 35600770 PMCID: PMC9120798 DOI: 10.1016/j.jgr.2021.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/09/2021] [Accepted: 10/06/2021] [Indexed: 02/06/2023] Open
Abstract
Background Gintonin-enriched fraction (GEF), a non-saponin fraction of ginseng, is a novel glycolipoprotein rich in hydrophobic amino acids. GEF has recently been shown to regulate lipid metabolism and browning in adipocytes; however, the mechanisms underlying its effects on energy metabolism and whether it affects sarcopenic obesity are unclear. We aimed to evaluate the effects of GEF on skeletal muscle atrophy in high-fat diet (HFD)-induced obese mice. Methods To examine the effect of GEF on sarcopenic obesity, 4-week-old male ICR mice were used. The mice were divided into four groups: chow diet (CD), HFD, HFD supplemented with 50 mg/kg/day GEF, or 150 mg/kg/day GEF for 6 weeks. We analyzed body mass gain and grip strength, histological staining, western blot analysis, and immunofluorescence to quantify changes in sarcopenic obesity-related factors. Results GEF inhibited body mass gain while HFD-fed mice gained 22.7 ± 2.0 g, whereas GEF-treated mice gained 14.3 ± 1.2 g for GEF50 and 11.8 ± 1.6 g for GEF150 by downregulating adipogenesis and inducing lipolysis and browning in white adipose tissue (WAT). GEF also enhanced mitochondrial biogenesis threefold in skeletal muscle. Furthermore, GEF-treated skeletal muscle exhibited decreased expression of muscle-specific atrophic genes, and promoted myogenic differentiation and increased muscle mass and strength in a dose-dependent manner (p < 0.05). Conclusion These findings indicate that GEF may have potential uses in preventing sarcopenic obesity by promoting energy expenditure and attenuating skeletal muscle atrophy.
Collapse
Affiliation(s)
- Heegu Jin
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam, Gyeonggi, Republic of Korea
| | - Hyun-Ji Oh
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam, Gyeonggi, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Boo-Yong Lee
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam, Gyeonggi, Republic of Korea
- Corresponding author. Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam, Gyeonggi, 13488, Republic of Korea.
| |
Collapse
|
44
|
Giroud M, Jodeleit H, Prentice KJ, Bartelt A. Adipocyte function and the development of cardiometabolic disease. J Physiol 2021; 600:1189-1208. [PMID: 34555180 DOI: 10.1113/jp281979] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/31/2021] [Indexed: 11/08/2022] Open
Abstract
Obesity is a medical disorder caused by multiple mechanisms of dysregulated energy balance. A major consequence of obesity is an increased risk to develop diabetes, diabetic complications and cardiovascular disease. While a better understanding of the molecular mechanisms linking obesity, insulin resistance and cardiovascular disease is needed, translational research of the human pathology is hampered by the available cellular and rodent model systems. Major barriers are the species-specific differences in energy balance, vascular biology and adipose tissue physiology, especially related to white and brown adipocytes, and adipose tissue browning. In rodents, non-shivering thermogenesis is responsible for a large part of energy expenditure, but humans possess much less thermogenic fat, which means temperature is an important variable in translational research. Mouse models with predisposition to dyslipidaemia housed at thermoneutrality and fed a high-fat diet more closely reflect human physiology. Also, adipocytes play a key role in the endocrine regulation of cardiovascular function. Adipocytes secrete a variety of hormones, lipid mediators and other metabolites that directly influence the local microenvironment as well as distant tissues. This is specifically apparent in perivascular depots, where adipocytes modulate vascular function and inflammation. Altogether, these mechanisms highlight the critical role of adipocytes in the development of cardiometabolic disease.
Collapse
Affiliation(s)
- Maude Giroud
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.,Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany
| | - Henrika Jodeleit
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Bavaria, Germany
| | - Kacey J Prentice
- Department of Molecular Metabolism & Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.,Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Bavaria, Germany.,Department of Molecular Metabolism & Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
45
|
Zwartjes MSZ, Gerdes VEA, Nieuwdorp M. The Role of Gut Microbiota and Its Produced Metabolites in Obesity, Dyslipidemia, Adipocyte Dysfunction, and Its Interventions. Metabolites 2021; 11:531. [PMID: 34436472 PMCID: PMC8398981 DOI: 10.3390/metabo11080531] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity is becoming an increasing problem worldwide and is often, but not invariably, associated with dyslipidemia. The gut microbiota is increasingly linked to cardiovascular disease, nonalcoholic fatty liver disease, and type 2 diabetes mellitus. However, relatively little focus has been attributed to the role of gut-microbiota-derived metabolites in the development of dyslipidemia and alterations in lipid metabolism. In this review, we discuss current data involved in these processes and point out the therapeutic potentials. We cover the ability of gut microbiota metabolites to alter lipoprotein lipase action, VLDL secretion, and plasma triglyceride levels, and its effects on reverse cholesterol transport, adipocyte dysfunction, and adipose tissue inflammation. Finally, the current intervention strategies for treatment of obesity and dyslipidemia is addressed with emphasis on the role of gut microbiota metabolites and its ability to predict treatment efficacies.
Collapse
Affiliation(s)
- Max S. Z. Zwartjes
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (V.E.A.G.); (M.N.)
- Department of Internal Medicine, Spaarne Gasthuis, Spaarnepoort 1, 2134 TM Hoofddorp, The Netherlands
| | - Victor E. A. Gerdes
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (V.E.A.G.); (M.N.)
- Department of Internal Medicine, Spaarne Gasthuis, Spaarnepoort 1, 2134 TM Hoofddorp, The Netherlands
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (V.E.A.G.); (M.N.)
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
- Department of Internal Medicine, Diabetes Center, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
46
|
Reyes-Farias M, Fos-Domenech J, Serra D, Herrero L, Sánchez-Infantes D. White adipose tissue dysfunction in obesity and aging. Biochem Pharmacol 2021; 192:114723. [PMID: 34364887 DOI: 10.1016/j.bcp.2021.114723] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/19/2022]
Abstract
Both obesity and aging are associated with the development of metabolic diseases such as type 2 diabetes and cardiovascular disease. Chronic low-grade inflammation of adipose tissue is one of the mechanisms implicated in the progression of these diseases. Obesity and aging trigger adipose tissue alterations that ultimately lead to a pro-inflammatory phenotype of the adipose tissue-resident immune cells. Obesity and aging also share other features such as a higher visceral vs. subcutaneous adipose tissue ratio and a decreased lifespan. Here, we review the common characteristics of obesity and aging and the alterations in white adipose tissue and resident immune cells. We focus on the adipose tissue metabolic derangements in obesity and aging such as inflammation and adipose tissue remodeling.
Collapse
Affiliation(s)
- Marjorie Reyes-Farias
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, Barcelona, Spain; Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - Julia Fos-Domenech
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain.
| | - David Sánchez-Infantes
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain; Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), E-28922 Madrid, Spain.
| |
Collapse
|
47
|
Liu F, He J, Liu B, Zhang P, Wang H, Sun X, Chu X, Guan W, Feng W, Bi Y, Zhu D. Association of Omental Adipocyte Hypertrophy and Fibrosis with Human Obesity and Type 2 Diabetes. Obesity (Silver Spring) 2021; 29:976-984. [PMID: 33943025 DOI: 10.1002/oby.23155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/04/2021] [Accepted: 02/07/2021] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Morphological alterations including adipocyte hypertrophy and fibrosis deposition are important surrogate markers of visceral adipose tissue function, but the relationships between these morphological changes and type 2 diabetes mellitus (T2DM) and impaired insulin sensitivity are poorly defined. METHODS Omental adipose tissue was obtained from 66 individuals with obesity but without T2DM (OB group), 93 individuals with both obesity and T2DM (T2DM group), and 15 individuals with normal BMI and normal glucose tolerance (NGT group). Adipocyte diameter and volume were measured through pathological section analysis. Pericellular and perilobular fibrosis was determined through picrosirius red staining and immunochemistry, while fibrosis-related genes were tested through gene expression and hydroxyproline content. RESULTS Compared with the NGT and OB groups, individuals from the T2DM group displayed increased adipocyte diameter and volume levels. Increased adipocyte size (diameter and volume) was positively associated with hyperglycemia and insulin resistance and inversely correlated with insulin sensitivity (using the Matsuda whole-body insulin sensitivity index assessment of insulin sensitivity) and β-cell function (disposition index 30 and disposition index 120). The fibrosis levels of the OB group were the highest out of the three groups, whereas the fibrosis levels of T2DM individuals were lower than the OB group but higher than the NGT group. Although fibrosis was negatively correlated with T2DM, fibrosis deposition was not remarkably associated with impaired systemic insulin sensitivity and glucose metabolism. CONCLUSIONS Compared with fibrosis deposition, adipocyte hypertrophy is more closely associated with T2DM and impaired systemic insulin sensitivity.
Collapse
Affiliation(s)
- Fangcen Liu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
- Department of Pathology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jielei He
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Boxun Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pengzi Zhang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Hongdong Wang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xitai Sun
- Department of General Surgery, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xuehui Chu
- Department of General Surgery, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenxian Guan
- Department of General Surgery, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenhuan Feng
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
48
|
Gater DR, Farkas GJ, Tiozzo E. Pathophysiology of Neurogenic Obesity After Spinal Cord Injury. Top Spinal Cord Inj Rehabil 2021; 27:1-10. [PMID: 33814879 PMCID: PMC7983633 DOI: 10.46292/sci20-00067] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Individuals with a spinal cord injury (SCI) have a unique physiology characterized by sarcopenia, neurogenic osteoporosis, neurogenic anabolic deficiency, sympathetic dysfunction, and blunted satiety associated with their SCI, all of which alter energy balance and subsequently body composition. The distinct properties of "neurogenic obesity" place this population at great risk for metabolic dysfunction, including systemic inflammation, hyperglycemia, dyslipidemia, and hypertension. The purpose of this article is to demonstrate the relationship between neurogenic obesity and the metabolic syndrome after SCI, highlighting the mechanisms associated with adipose tissue pathology and those respective comorbidities. Additionally, representative studies of persons with SCI will be provided to elucidate the severity of the problem and to prompt greater vigilance among SCI specialists as well as primary care providers in order to better manage the epidemic from a public health perspective.
Collapse
Affiliation(s)
- David R. Gater
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Gary J. Farkas
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida
| | - Eduard Tiozzo
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
49
|
Variables Associated with Short-Term Weight Loss in a Cohort of Patients with Morbid Obesity According to Age and Three Types of Bariatric Surgery. J Clin Med 2020; 9:jcm9113537. [PMID: 33147793 PMCID: PMC7692348 DOI: 10.3390/jcm9113537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 11/26/2022] Open
Abstract
Background The percentage of excess weight lost (%EWL) after bariatric surgery (BS) shows great discrepancies from one individual to another. Objective To evaluate the %EWL one year after BS and to determine the existence of baseline biomarkers associated with weight loss. Methods We studied 329 patients with morbid obesity undergoing three types of BS (biliopancreatic diversion (BPD), Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG)), depending on the %EWL one year after surgery: good responders (GR) (%EWL ≥ 50%) and non-responders (NR) (%EWL < 50%). Results The GR presented a higher percentage of change in anthropometric and biochemical variables compared to the NR group, even within each type of BS. There was a greater percentage of GR among those who underwent RYGB. The patients who underwent SG showed the lowest decrease in biochemical variables, both in GR and NR. Within the GR group, those with a lower age showed greater improvement compared to the other age groups. A %EWL ≥50% was negatively associated with the age and atherogenic index of plasma (AIP), and positively with the type of BS (RYGB). Conclusions The GR group was associated with lower age and AIP and undergoing RYGB. Additionally, those patients who underwent SG showed a lower metabolic improvement.
Collapse
|