1
|
Drewa J, Lazar-Juszczak K, Adamowicz J, Juszczak K. Periprostatic Adipose Tissue as a Contributor to Prostate Cancer Pathogenesis: A Narrative Review. Cancers (Basel) 2025; 17:372. [PMID: 39941741 PMCID: PMC11816168 DOI: 10.3390/cancers17030372] [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: 12/17/2024] [Revised: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Periprostatic adipose tissue (PPAT) contributes to the pathogenesis of prostate cancer. The purpose of this study was to review and summarize the literature on the role of PPAT in prostate cancer pathogenesis. Moreover, we evaluated the clinical implication of PPAT in patients with prostate cancer. We performed a scoping literature review of PubMed from January 2002 to November 2024. Search terms included "periprostatic adipose tissue", "adipokines", and "prostate cancer". Secondary search involved reference lists of eligible articles. The key criterion was to identify studies that included PPAT, adipokines, and their role in prostate cancer biology and clinical features. In total 225 publications were selected for inclusion in this review. The studies contained in publications allowed us to summarize the data on the pathogenesis of PPAT as a contributor to prostate cancer biology and its aggressiveness. The review also presents new research directions for PPAT as a new target for the treatment of prostate cancer. Based on the current review, it can be stated that PPAT plays an important role in prostate cancer pathogenesis. Moreover, PPAT seems to be a promising target point when it comes to finding new therapies in patients with more aggressive and/or advanced stages of prostate cancer.
Collapse
Affiliation(s)
- Julia Drewa
- Department of Urology and Andrology, Collegium Medicum, Nicolaus Copernicus University, 85-094 Bydgoszcz, Poland
| | - Katarzyna Lazar-Juszczak
- Primary Health Care Clinic of the Ujastek Medical Center, 31-752 Cracow, Poland
- Krakow University of Health Promotion, 31-158 Cracow, Poland
| | - Jan Adamowicz
- Department of Urology and Andrology, Collegium Medicum, Nicolaus Copernicus University, 85-094 Bydgoszcz, Poland
- Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, 85-094 Bydgoszcz, Poland
| | - Kajetan Juszczak
- Department of Urology and Andrology, Collegium Medicum, Nicolaus Copernicus University, 85-094 Bydgoszcz, Poland
| |
Collapse
|
2
|
Wang Y, Bo Y, Liu Y, Zhou J, Nguyen D, Baskaran D, Liu Y, Wang H. Metabolic labeling and targeted modulation of adipocytes. Biomater Sci 2025; 13:434-445. [PMID: 39648977 PMCID: PMC11758917 DOI: 10.1039/d4bm01352b] [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] [Indexed: 12/10/2024]
Abstract
Adipocytes play a critical role in energy storage and endocrine signaling and are associated with various diseases such as cancer and diabetes. Facile strategies to engineer adipocytes have long been pursued for elucidating adipocyte biology and developing adipocyte-based therapies. Herein, we report metabolic glycan labeling of adipocytes and subsequent targeted modulation of adipocytes via click chemistry. We show that azido tags expressed on the surface of adipocytes can persist for over 4 days. By conjugating dibenzocyclooctyne (DBCO)-cargos onto azido-labeled adipocytes via click chemistry, the cargos can be retained on the adipocyte membrane for over 12 hours. We further show that signaling molecules including adiponectin, calreticulin, mannose-binding lectin 2, and milk fat globule-EGF factor 8 protein can be conjugated to adipocytes to orchestrate their phagocytosis by macrophages. The azido-labeled adipocytes grafted into mice can also mediate targeted conjugation of DBCO-cargos in vivo. This adipocyte labeling and targeting technology will facilitate the development of adipocyte-based therapies and provides a new platform for manipulating the interaction between adipocytes and other types of cells.
Collapse
Affiliation(s)
- Yueji Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yang Bo
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Yusheng Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Jiadiao Zhou
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Daniel Nguyen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Dhyanesh Baskaran
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Yuan Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
- Cancer Center at Illinois (CCIL), Urbana, IL 61801, USA
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
3
|
Lee HY, Min KJ. Dietary Restriction and Lipid Metabolism: Unveiling Pathways to Extended Healthspan. Nutrients 2024; 16:4424. [PMID: 39771045 PMCID: PMC11678862 DOI: 10.3390/nu16244424] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Dietary restriction (DR) has been reported to be a significant intervention that influences lipid metabolism and potentially modulates the aging process in a wide range of organisms. Lipid metabolism plays a pivotal role in the regulation of aging and longevity. In this review, we summarize studies on the significant role of lipid metabolism in aging in relation to DR. As a potent intervention to slow down aging, DR has demonstrated promising effects on lipid metabolism, influencing the aging processes across various species. The current review focuses on the relationships among DR-related molecular signaling proteins such as the sirtuins, signaling pathways such as the target of rapamycin and the insulin/insulin-like growth factor (IGF)-1, lipid metabolism, and aging. Furthermore, the review presents research results on diet-associated changes in cell membrane lipids and alterations in lipid metabolism caused by commensal bacteria, highlighting the importance of lipid metabolism in aging. Overall, the review explores the interplay between diet, lipid metabolism, and aging, while presenting untapped areas for further understanding of the aging process.
Collapse
Affiliation(s)
| | - Kyung-Jin Min
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea;
| |
Collapse
|
4
|
Bakondi E, Jung T, Marg S, Schnell V, Weber D, Schulz TJ, Grune T, Höhn A. Palmitic acid and eicosapentaenoic acid supplementation in 3T3 adipocytes: impact on lipid storage and oxidative stress. Redox Rep 2024; 29:2430882. [PMID: 39607809 DOI: 10.1080/13510002.2024.2430882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
OBJECTIVES Obesity is a worldwide public health problem, predisposing individuals to serious cardiovascular and metabolic complications such as type 2 diabetes mellitus. White adipose tissue serves as an important regulator of energy balance, and its expansion in obesity can trigger inflammatory reactions and oxidative stress, which can also lead to insulin resistance. Adipocytes, with a key role in regulating metabolic homeostasis, respond to increased calorie intake and altered fatty acid composition with hypertrophy or hyperplasia. Of particular interest are saturated fatty acids such as palmitic acid and omega-3 polyunsaturated fatty acids such as eicosapentaenoic acid (EPA), which have differential effects on adipocyte function and inflammation. METHODS Using 3T3-L1 cells as a model for adipocytes, we evaluated the effects of PA and EPA on lipid accumulation, droplet size, and oxidative stress markers. RESULTS We were able to show that EPA supplementation in 3T3 adipocytes does not lead to excessive lipid accumulation, but rather reduces the size of lipid droplets and also induces redox changes due to the unsaturated nature of EPA. DISCUSSION These results emphasize the contrasting roles of PA and EPA and the importance of fatty acid composition in the regulation of adipocyte function.
Collapse
Affiliation(s)
- Edina Bakondi
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Tobias Jung
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Susanna Marg
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Vanessa Schnell
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Daniela Weber
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Annika Höhn
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
5
|
Assaf S, Park J, Chowdhry N, Ganapuram M, Mattathil S, Alakeel R, Kelly OJ. Unraveling the Evolutionary Diet Mismatch and Its Contribution to the Deterioration of Body Composition. Metabolites 2024; 14:379. [PMID: 39057702 PMCID: PMC11279030 DOI: 10.3390/metabo14070379] [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: 06/01/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Over the millennia, patterns of food consumption have changed; however, foods were always whole foods. Ultra-processed foods (UPFs) have been a very recent development and have become the primary food source for many people. The purpose of this review is to propose the hypothesis that, forsaking the evolutionary dietary environment, and its complex milieu of compounds resulting in an extensive metabolome, contributes to chronic disease in modern humans. This evolutionary metabolome may have contributed to the success of early hominins. This hypothesis is based on the following assumptions: (1) whole foods promote health, (2) essential nutrients cannot explain all the benefits of whole foods, (3) UPFs are much lower in phytonutrients and other compounds compared to whole foods, and (4) evolutionary diets contributed to a more diverse metabolome. Evidence will be presented to support this hypothesis. Nutrition is a matter of systems biology, and investigating the evolutionary metabolome, as compared to the metabolome of modern humans, will help elucidate the hidden connections between diet and health. The effect of the diet on the metabolome may also help shape future dietary guidelines, and help define healthy foods.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Owen J. Kelly
- College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA; (S.A.); (J.P.); (N.C.); (M.G.); (S.M.); (R.A.)
| |
Collapse
|
6
|
Blade SP, Falkowski DJ, Bachand SN, Pagano SJ, Chin L. Mechanobiology of Adipocytes. BIOLOGY 2024; 13:434. [PMID: 38927314 PMCID: PMC11200640 DOI: 10.3390/biology13060434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024]
Abstract
The growing obesity epidemic necessitates increased research on adipocyte and adipose tissue function and disease mechanisms that progress obesity. Historically, adipocytes were viewed simply as storage for excess energy. However, recent studies have demonstrated that adipocytes play a critical role in whole-body homeostasis, are involved in cell communication, experience forces in vivo, and respond to mechanical stimuli. Changes to the adipocyte mechanical microenvironment can affect function and, in some cases, contribute to disease. The aim of this review is to summarize the current literature on the mechanobiology of adipocytes. We reviewed over 100 papers on how mechanical stress is sensed by the adipocyte, the effects on cell behavior, and the use of cell culture scaffolds, particularly those with tunable stiffness, to study adipocyte behavior, adipose cell and tissue mechanical properties, and computational models. From our review, we conclude that adipocytes are responsive to mechanical stimuli, cell function and adipogenesis can be dictated by the mechanical environment, the measurement of mechanical properties is highly dependent on testing methods, and current modeling practices use many different approaches to recapitulate the complex behavior of adipocytes and adipose tissue. This review is intended to aid future studies by summarizing the current literature on adipocyte mechanobiology.
Collapse
Affiliation(s)
- Sean P. Blade
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Dylan J. Falkowski
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Sarah N. Bachand
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Steven J. Pagano
- Department of Mechanical Engineering, Widener University, Chester, PA 19013, USA;
| | - LiKang Chin
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| |
Collapse
|
7
|
Kang DW, Koh SH, Kim MK, Kim DY. Effects of aquatic versus land-based exercise on irisin and fibroblast growth factor 21 expression and triiodothyronine and free fatty acid levels in elderly women. Osong Public Health Res Perspect 2024; 15:238-247. [PMID: 38988027 PMCID: PMC11237314 DOI: 10.24171/j.phrp.2023.0394] [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: 01/03/2024] [Accepted: 03/06/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND This study investigated the impacts of exercise on irisin and fibroblast growth factor 21 (FGF-21) expression, as well as triiodothyronine (T3 ) and free fatty acid (FFA) levels in elderly women. METHODS Thirty women aged 65 to 70 years (10 per group) were randomly assigned to aquatic exercise, land exercise, and control groups. The aquatic and land groups engaged in 3 exercise sessions per week (60 min/session) for 16 weeks. The intensity was progressively increased every 4 weeks. RESULTS Irisin and FGF-21 levels significantly increased in the aquatic exercise group. In the posttest, the aquatic exercise group had the highest irisin levels. Significant findings were observed for irisin and FGF-21 for the main effect between aquatic and band exercise groups (p<0.05 for both), the main effect between measurement times (p<0.01 and p<0.001, respectively), and the interaction effect (p<0.05 and p<0.001, respectively). The irisin level was significantly higher in the aquatic than in the land group 30 minutes after the last session (p<0.05). In both exercise groups, T3 levels were significantly higher 30 minutes after the final session (p<0.05) than before the program. The FFA level was significantly higher in the aquatic exercise group than the others. In the aquatic group, FFA levels were significantly higher 30 minutes after both the first (p<0.01) and the last (p<0.001) session compared to pre-program values. CONCLUSION Differences in exercise type and environment can promote fat metabolism by stimulating hormonal changes that induce brown fat activity and browning.
Collapse
Affiliation(s)
- Du-Wang Kang
- Department of Physical Education, Pusan National University, Busan, Republic of Korea
| | - Su-Han Koh
- Department of Physical Education, Pusan National University, Busan, Republic of Korea
| | - Min-Kyo Kim
- Department of Physical Education, Pusan National University, Busan, Republic of Korea
| | - Do-Yeon Kim
- Department of Physical Education, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
8
|
Nagagata BA, Brito G, Ornellas F, Mandarim-de-Lacerda CA, Aguila MB. Melatonin supplementation in obese mothers reduces hypothalamic inflammation and enhances thermogenesis in mice progeny. J Nutr Biochem 2024; 128:109625. [PMID: 38521130 DOI: 10.1016/j.jnutbio.2024.109625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/07/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
Maternal obesity might induce obesity and metabolic alterations in the progeny. The study aimed to determine the effect of supplementing obese mothers with Mel (Mel) on thermogenesis and inflammation. C57BL/6 female mice (mothers) were fed from weaning to 12 weeks control diet (C, 17% kJ as fat) or a high-fat diet (HF, 49% kJ as fat) and then matted with male mice fed the control diet. Melatonin (10 mg/kg daily) was supplemented to mothers during gestation and lactation, forming the groups C, CMel, HF, and HFMel (n = 10/group). Twelve-week male offspring were studied (plasma biochemistry, immunohistochemistry, protein, and gene expressions at the hypothalamus - Hyp, subcutaneous white adipose tissue - sWAT, and interscapular brown adipose tissue - iBAT). Comparing HFMel vs. HF offspring, fat deposits and plasmatic proinflammatory markers decreased. Also, HFMel showed decreased Hyp proinflammatory markers and neuropeptide Y (anabolic) expression but improved proopiomelanocortin (catabolic) expression. Besides, HFMel sWAT adipocytes changed to a beige phenotype with-beta-3 adrenergic receptor and uncoupling protein-1 activation, concomitant with browning genes activation, triggering the iBAT thermogenic activity. In conclusion, compelling evidence indicated the beneficial effects of supplementing obese mothers with Mel on the health of their mature male offspring. Mel led to sWAT browning-related gene enhancement, increased iBAT thermogenis, and mitigated hypothalamic inflammation. Also, principal component analysis of the data significantly separated the untreated obese mother progeny from the progeny of treated obese mothers. If confirmed in humans, the findings encourage a future guideline recommending Mel supplementation during pregnancy and breastfeeding.
Collapse
Affiliation(s)
- Brenda A Nagagata
- Metabolism section, Laboratory of Morphometry, Metabolism and Cardiovascular Disease, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Nutrition section, Laboratory of Morphometry, Metabolism and Cardiovascular Disease, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabrielle Brito
- Metabolism section, Laboratory of Morphometry, Metabolism and Cardiovascular Disease, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Di Lorenzo M, Cacciapuoti N, Lonardo MS, Nasti G, Gautiero C, Belfiore A, Guida B, Chiurazzi M. Pathophysiology and Nutritional Approaches in Polycystic Ovary Syndrome (PCOS): A Comprehensive Review. Curr Nutr Rep 2023; 12:527-544. [PMID: 37213054 PMCID: PMC10444658 DOI: 10.1007/s13668-023-00479-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2023] [Indexed: 05/23/2023]
Abstract
PURPOSE OF REVIEW Polycystic ovary syndrome (PCOS) is the most common endocrine and metabolic disorder in women of reproductive age worldwide. This disease causes menstrual, metabolic, and biochemical abnormalities such as hyperandrogenism, oligo-anovulatory menstrual cycles, polycystic ovary, hyperleptinemia, insulin resistance (IR), and cardiometabolic disorders, often associated with overweight or obesity and visceral adiposity. RECENT FINDINGS The etiology and pathophysiology of PCOS are not yet fully understood, but insulin seems to play a key role in this disease. PCOS shares an inflammatory state with other chronic diseases such as obesity, type II diabetes, and cardiovascular diseases; however, recent studies have shown that a healthy nutritional approach can improve IR and metabolic and reproductive functions, representing a valid therapeutic strategy to ameliorate PCOS symptomatology. This review aimed to summarize and collect evidence about different nutritional approaches such as the Mediterranean diet (MedDiet) and the ketogenic diet (KD), as well as bariatric surgery and nutraceutical supplementation as probiotics, prebiotics, and synbiotics, among the others, used in patients with PCOS.
Collapse
Affiliation(s)
- M Di Lorenzo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
- Infectious Diseases and Gender Medicine Unit, Cotugno Hospital, AO Dei Colli, Naples, Italy
| | - N Cacciapuoti
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - M S Lonardo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - G Nasti
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - C Gautiero
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - A Belfiore
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - B Guida
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - M Chiurazzi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy.
- Department of Medical Oncology, AO "A. Cardarelli", Naples, Italy.
| |
Collapse
|
10
|
Ochiai M, Fierstein S, XsSali F, DeVito N, Purkey LR, May R, Correa-Medina A, Kelley M, Page TD, DeCicco-Skinner K. Unlocking Drug Resistance in Multiple Myeloma: Adipocytes as Modulators of Treatment Response. Cancers (Basel) 2023; 15:4347. [PMID: 37686623 PMCID: PMC10486466 DOI: 10.3390/cancers15174347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/13/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Multiple myeloma (MM) is an incurable hematological malignancy characterized by the clonal proliferation of malignant plasma cells. Despite the development of a diverse array of targeted drug therapies over the last decade, patients often relapse and develop refractory disease due to multidrug resistance. Obesity is a growing public health threat and a risk factor for multiple myeloma, although the mechanisms by which obesity contributes to MM growth and progression have not been fully elucidated. In the present study, we evaluated whether crosstalk between adipocytes and MM cells promoted drug resistance and whether this was amplified by obesity. Human adipose-derived stem cells (ASCs) from nineteen normal (BMI = 20-25 kg/m2), overweight (25-30 kg/m2), or obese (30-35 kg/m2) patients undergoing elective liposuction were utilized. Cells were differentiated into adipocytes, co-cultured with RPMI 8226 or U266B1 multiple myeloma cell lines, and treated with standard MM therapies, including bortezomib or a triple combination of bortezomib, dexamethasone, and lenalidomide. We found that adipocytes from overweight and obese individuals increased cell adhesion-mediated drug resistance (CAM-DR) survival signals in MM cells, and P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP) drug transporter expression. Further, co-culture enhanced in vitro angiogenesis, MMP-2 activity, and protected MM cells from drug-induced decreases in viability. In summary, we provide an underlying mechanism by which obesity can impair the drug response to MM and allow for recurrence and/or disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kathleen DeCicco-Skinner
- Department of Biology, American University, 4400 Massachusetts Ave, NW, Washington, DC 20016, USA
| |
Collapse
|
11
|
Knaack DA, Chang J, Thomas MJ, Sorci-Thomas MG, Chen Y, Sahoo D. Scavenger receptor class B type I is required for efficient glucose uptake and metabolic homeostasis in adipocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.554190. [PMID: 37662321 PMCID: PMC10473602 DOI: 10.1101/2023.08.21.554190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Obesity is a worldwide epidemic and places individuals at a higher risk for developing comorbidities that include cardiovascular disease and type 2 diabetes. Adipose tissue contains adipocytes that are responsible for lipid metabolism and reducing misdirected lipid storage. Adipocytes facilitate this process through insulin-mediated uptake of glucose and its subsequent metabolism into triglycerides for storage. During obesity, adipocytes become insulin resistant and have a reduced ability to mediate glucose import, thus resulting in whole-body metabolic dysfunction. Scavenger receptor class B type I (SR-BI) has been implicated in glucose uptake in skeletal muscle and adipocytes via its native ligands, apolipoprotein A-1 and high-density lipoproteins. Further, SR-BI translocation to the cell surface in adipocytes is sensitive to insulin stimulation. Using adipocytes differentiated from ear mesenchymal stem cells isolated from wild-type and SR-BI knockout (SR-BI -/- ) mice as our model system, we tested the hypothesis that SR-BI is required for insulin-mediated glucose uptake and regulation of energy balance in adipocytes. We demonstrated that loss of SR-BI in adipocytes resulted in inefficient glucose uptake regardless of cell surface expression levels of glucose transporter 4 compared to WT adipocytes. We also observed reduced glycolytic capacity, increased lipid biosynthesis, and dysregulated expression of lipid metabolism genes in SR-BI -/- adipocytes compared to WT adipocytes. These results partially support our hypothesis and suggest a novel role for SR-BI in glucose uptake and metabolic homeostasis in adipocytes.
Collapse
|
12
|
Amin A, Badenes M, Tüshaus J, de Carvalho É, Burbridge E, Faísca P, Trávníčková K, Barros A, Carobbio S, Domingos PM, Vidal-Puig A, Moita LF, Maguire S, Stříšovský K, Ortega FJ, Fernández-Real JM, Lichtenthaler SF, Adrain C. Semaphorin 4B is an ADAM17-cleaved adipokine that inhibits adipocyte differentiation and thermogenesis. Mol Metab 2023; 73:101731. [PMID: 37121509 PMCID: PMC10197113 DOI: 10.1016/j.molmet.2023.101731] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/02/2023] Open
Abstract
OBJECTIVE The metalloprotease ADAM17 (also called TACE) plays fundamental roles in homeostasis by shedding key signaling molecules from the cell surface. Although its importance for the immune system and epithelial tissues is well-documented, little is known about the role of ADAM17 in metabolic homeostasis. The purpose of this study was to determine the impact of ADAM17 expression, specifically in adipose tissues, on metabolic homeostasis. METHODS We used histopathology, molecular, proteomic, transcriptomic, in vivo integrative physiological and ex vivo biochemical approaches to determine the impact of adipose tissue-specific deletion of ADAM17 upon adipocyte and whole organism metabolic physiology. RESULTS ADAM17adipoq-creΔ/Δ mice exhibited a hypermetabolic phenotype characterized by elevated energy consumption and increased levels of adipocyte thermogenic gene expression. On a high fat diet, these mice were more thermogenic, while exhibiting elevated expression levels of genes associated with lipid oxidation and lipolysis. This hypermetabolic phenotype protected mutant mice from obesogenic challenge, limiting weight gain, hepatosteatosis and insulin resistance. Activation of beta-adrenoceptors by the neurotransmitter norepinephrine, a key regulator of adipocyte physiology, triggered the shedding of ADAM17 substrates, and regulated ADAM17 expression at the mRNA and protein levels, hence identifying a functional connection between thermogenic licensing and the regulation of ADAM17. Proteomic studies identified Semaphorin 4B (SEMA4B), as a novel ADAM17-shed adipokine, whose expression is regulated by physiological thermogenic cues, that acts to inhibit adipocyte differentiation and dampen thermogenic responses in adipocytes. Transcriptomic data showed that cleaved SEMA4B acts in an autocrine manner in brown adipocytes to repress the expression of genes involved in adipogenesis, thermogenesis, and lipid uptake, storage and catabolism. CONCLUSIONS Our findings identify a novel ADAM17-dependent axis, regulated by beta-adrenoceptors and mediated by the ADAM17-cleaved form of SEMA4B, that modulates energy balance in adipocytes by inhibiting adipocyte differentiation, thermogenesis and lipid catabolism.
Collapse
Affiliation(s)
- Abdulbasit Amin
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Nigeria
| | - Marina Badenes
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Faculty of Veterinary Medicine, Lusofona University, Lisbon, Portugal; Faculty of Veterinary Nursing, Polytechnic Institute of Lusofonia, Lisbon, Portugal
| | - Johanna Tüshaus
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Érika de Carvalho
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Instituto de Tecnologia Química da Universidade Nova de Lisboa (ITQB-Nova), Oeiras, Portugal
| | - Emma Burbridge
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, N. Ireland
| | - Pedro Faísca
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
| | - Květa Trávníčková
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - André Barros
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
| | - Stefania Carobbio
- Centro de Investigacíon Principe Felipe (CIPF), Valencia, Spain; Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, UK
| | - Pedro M Domingos
- Instituto de Tecnologia Química da Universidade Nova de Lisboa (ITQB-Nova), Oeiras, Portugal
| | - Antonio Vidal-Puig
- Centro de Investigacíon Principe Felipe (CIPF), Valencia, Spain; Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, UK
| | - Luís F Moita
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
| | - Sarah Maguire
- Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, N. Ireland
| | - Kvido Stříšovský
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Francisco J Ortega
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain; Department of Medical Sciences, University of Girona, Girona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), and Institute of Salud Carlos III (ISCIII), Madrid, Spain
| | - José Manuel Fernández-Real
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain; Department of Medical Sciences, University of Girona, Girona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), and Institute of Salud Carlos III (ISCIII), Madrid, Spain
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Colin Adrain
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, N. Ireland.
| |
Collapse
|
13
|
Vourdoumpa A, Paltoglou G, Charmandari E. The Genetic Basis of Childhood Obesity: A Systematic Review. Nutrients 2023; 15:1416. [PMID: 36986146 PMCID: PMC10058966 DOI: 10.3390/nu15061416] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/05/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Overweight and obesity in childhood and adolescence represents one of the most challenging public health problems of our century owing to its epidemic proportions and the associated significant morbidity, mortality, and increase in public health costs. The pathogenesis of polygenic obesity is multifactorial and is due to the interaction among genetic, epigenetic, and environmental factors. More than 1100 independent genetic loci associated with obesity traits have been currently identified, and there is great interest in the decoding of their biological functions and the gene-environment interaction. The present study aimed to systematically review the scientific evidence and to explore the relation of single-nucleotide polymorphisms (SNPs) and copy number variants (CNVs) with changes in body mass index (BMI) and other measures of body composition in children and adolescents with obesity, as well as their response to lifestyle interventions. Twenty-seven studies were included in the qualitative synthesis, which consisted of 7928 overweight/obese children and adolescents at different stages of pubertal development who underwent multidisciplinary management. The effect of polymorphisms in 92 different genes was assessed and revealed SNPs in 24 genetic loci significantly associated with BMI and/or body composition change, which contribute to the complex metabolic imbalance of obesity, including the regulation of appetite and energy balance, the homeostasis of glucose, lipid, and adipose tissue, as well as their interactions. The decoding of the genetic and molecular/cellular pathophysiology of obesity and the gene-environment interactions, alongside with the individual genotype, will enable us to design targeted and personalized preventive and management interventions for obesity early in life.
Collapse
Affiliation(s)
- Aikaterini Vourdoumpa
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
| | - George Paltoglou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
- Division of Endocrinology and Metabolism, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
14
|
Inhibitory Effects of Loganin on Adipogenesis In Vitro and In Vivo. Int J Mol Sci 2023; 24:ijms24054752. [PMID: 36902181 PMCID: PMC10003152 DOI: 10.3390/ijms24054752] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/18/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Obesity is characterized by the excessive accumulation of mature adipocytes that store surplus energy in the form of lipids. In this study, we investigated the inhibitory effects of loganin on adipogenesis in mouse preadipocyte 3T3-L1 cells and primary cultured adipose-derived stem cells (ADSCs) in vitro and in mice with ovariectomy (OVX)- and high-fat diet (HFD)-induced obesity in vivo. For an in vitro study, loganin was co-incubated during adipogenesis in both 3T3-L1 cells and ADSCs, lipid droplets were evaluated by oil red O staining, and adipogenesis-related factors were assessed by qRT-PCR. For in vivo studies, mouse models of OVX- and HFD-induced obesity were orally administered with loganin, body weight was measured, and hepatic steatosis and development of excessive fat were evaluated by histological analysis. Loganin treatment reduced adipocyte differentiation by accumulating lipid droplets through the downregulation of adipogenesis-related factors, including peroxisome proliferator-activated receptor γ (Pparg), CCAAT/enhancer-binding protein α (Cebpa), perilipin 2 (Plin2), fatty acid synthase (Fasn), and sterol regulatory element binding transcription protein 1 (Srebp1). Loganin administration prevented weight gain in mouse models of obesity induced by OVX and HFD. Further, loganin inhibited metabolic abnormalities, such as hepatic steatosis and adipocyte enlargement, and increased the serum levels of leptin and insulin in both OVX- and HFD-induced obesity models. These results suggest that loganin is a potential candidate for preventing and treating obesity.
Collapse
|
15
|
Kim YI, Lee ES, Song EJ, Shin DU, Eom JE, Shin HS, Kim JE, Oh JY, Nam YD, Lee SY. Lacticaseibacillus paracasei AO356 ameliorates obesity by regulating adipogenesis and thermogenesis in C57BL/6J male mice. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
16
|
Ilieva M, Uchida S. Potential Involvement of LncRNAs in Cardiometabolic Diseases. Genes (Basel) 2023; 14:213. [PMID: 36672953 PMCID: PMC9858747 DOI: 10.3390/genes14010213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
Characterized by cardiovascular disease and diabetes, cardiometabolic diseases are a major cause of mortality around the world. As such, there is an urgent need to understand the pathogenesis of cardiometabolic diseases. Increasing evidence suggests that most of the mammalian genome are transcribed as RNA, but only a few percent of them encode for proteins. All of the RNAs that do not encode for proteins are collectively called non-protein-coding RNAs (ncRNAs). Among these ncRNAs, long ncRNAs (lncRNAs) are considered as missing keys to understand the pathogeneses of various diseases, including cardiometabolic diseases. Given the increased interest in lncRNAs, in this study, we will summarize the latest trend in the lncRNA research from the perspective of cardiometabolism and disease by focusing on the major risk factors of cardiometabolic diseases: obesity, cholesterol, diabetes, and hypertension. Because genetic inheritance is unavoidable in cardiometabolic diseases, we paid special attention to the genetic factors of lncRNAs that may influence cardiometabolic diseases.
Collapse
Affiliation(s)
| | - Shizuka Uchida
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, DK-2450 Copenhagen SV, Denmark or
| |
Collapse
|
17
|
Kelvin Barros Dias K, Lima Cardoso A, Alice Farias da Costa A, Fonseca Passos M, Emmerson Ferreira da Costa C, Narciso da Rocha Filho G, Helena de Aguiar Andrade E, Luque R, Adriano Santos do Nascimento L, Coelho Rodrigues Noronha R. Biological activities from andiroba (Carapa guianensis Aublet.) and its biotechnological applications: a systematic review. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
18
|
Ma J, Sui F, Liu Y, Yuan M, Dang H, Liu R, Shi B, Hou P. Sorafenib decreases glycemia by impairing hepatic glucose metabolism. Endocrine 2022; 78:446-457. [PMID: 36205915 DOI: 10.1007/s12020-022-03202-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/17/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE Sorafenib has been reported to reduce blood glucose levels in diabetic and non-diabetic patients in previous retrospective studies. However, the mechanism of which the hypoglycemic effects of sorafenib is not clearly explored. In this study, we investigated the effect of sorafenib on blood glucose levels in diabetic and normal mice and explored the possible mechanism. METHODS We established a mouse model of type 2 diabetes by a high-fat diet combined with a low-dose of streptozotocin (STZ), to identify the hypoglycemic effect of sorafenib in different mice. Glucose tolerance, insulin tolerance and pyruvate tolerance tests were done after daily gavage with sorafenib to diabetic and control mice. To explore the molecular mechanism by which sorafenib regulates blood glucose levels, hepatic glucose metabolism signaling was studied by a series of in vivo and in vitro experiments. RESULTS Sorafenib reduced blood glucose levels in both control and diabetic mice, particularly in the latter. The diabetic mice exhibited improved glucose and insulin tolerance after sorafenib treatment. Further studies showed that the expressions of gluconeogenesis-related enzymes, such as PCK1, G6PC and PCB, were significantly decreased upon sorafenib treatment. Mechanistically, sorafenib downregulates the expression of c-MYC downstream targets PCK1, G6PC and PCB through blocking the ERK/c-MYC signaling pathway, thereby playing its hypoglycemic effect by impairing hepatic glucose metabolism. CONCLUSION Sorafenib reduces blood glucose levels through downregulating gluconeogenic genes, especially in diabetic mice, suggesting the patients with T2DM when treated with sorafenib need more emphasis in monitoring blood glucose to avoid unnecessary hypoglycemia.
Collapse
Affiliation(s)
- Jingjing Ma
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, P.R. China
| | - Fang Sui
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, P.R. China
| | - Yan Liu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, P.R. China
| | - Mengmeng Yuan
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, P.R. China
| | - Hui Dang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, P.R. China
| | - Rui Liu
- Department of Radio-Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, P.R. China
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, P.R. China
| | - Peng Hou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, P.R. China.
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province and Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, P.R. China.
| |
Collapse
|
19
|
Dou X, Mao T, Ma Y, Jia F, Liu Y, Liu X. Fibrotic and inflammatory characteristics of epidural fat adjacent to the ossification area in patients with ossification of the ligament flavum. JOR Spine 2022; 5:e1229. [PMID: 36601380 PMCID: PMC9799088 DOI: 10.1002/jsp2.1229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 09/20/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Objectives To observe histological and inflammatory characteristics of epidural fat (EF) adjacent to the ossification area in patients with ossification of the thoracic ligament flavum (TOLF) and provide a preliminary research basis for investigating the impact of the EF on OLF. Methods Samples of EF and autologous subcutaneous adipose tissue (SCAT) from TOLF patients (n = 26) and non-TOLF patients (n = 23) were harvested during posterior thoracic spine surgery. Adipocyte size and fibrosis were measured by histology. Vascularization and inflammatory cell infiltration were evaluated by immunohistochemical staining. Adipogenesis-related genes were assessed by real-time quantitative PCR. Conditioned media from cultured EF were evaluated via enzyme-linked immunosorbent assay to detect the secretion of inflammatory cytokines, including interleukin-6 (IL-6), tumor necrosis factor (TNF-α), and leptin. The phosphorylated STAT3 protein level in ligament flavum (LF) was examined using Western blot. Results Adipocytes size in EF was similar between in the TOLF and non-TOLF groups, but significantly smaller than that from autologous SCAT. Adipogenesis-related mRNA expression in EF was lower than that in SCAT. More fibrosis and vascularization were found in TOLF than in non-TOLF. EF in the TOLF group exhibited more macrophages and B lymphocytes infiltrated. The levels of cytokines such as IL-6, TNF-α, and leptin secreted by EF were significantly higher in the TOLF group than non-TOLF. The level of phosphorylated STAT3 in LF was significantly upregulated in the TOLF group. Conclusions Morphologically, EF adjacent to the ossification area is smaller and more uniform than autologous SCAT, exhibiting a characteristic similarity to visceral fat. EF in the TOLF group shows a more fibrotic, vascularized, and inflammatory phenotype, which secretes multiple cytokines. The phosphorylated STAT3 protein was significantly upregulated in the TOLF group. Whether these properties of EF directly affect the process of OLF needs to be further explored.
Collapse
Affiliation(s)
- Xinyu Dou
- Department of OrthopedicsPeking University Third HospitalBeijingChina
- Beijing Key Laboratory of Spinal DiseasesBeijingChina
- Engineering Research Center of Bone and Joint Precision MedicineBeijingChina
| | - Tianli Mao
- Department of OrthopedicsPeking University Third HospitalBeijingChina
- Beijing Key Laboratory of Spinal DiseasesBeijingChina
- Engineering Research Center of Bone and Joint Precision MedicineBeijingChina
| | - Yunlong Ma
- Pain Medicine CenterPeking University Third HospitalBeijingChina
| | - Fei Jia
- Department of OrthopedicsPeking University Third HospitalBeijingChina
- Beijing Key Laboratory of Spinal DiseasesBeijingChina
- Engineering Research Center of Bone and Joint Precision MedicineBeijingChina
| | - Yu Liu
- Department of OrthopedicsPeking University Third HospitalBeijingChina
- Beijing Key Laboratory of Spinal DiseasesBeijingChina
- Engineering Research Center of Bone and Joint Precision MedicineBeijingChina
| | - Xiaoguang Liu
- Department of OrthopedicsPeking University Third HospitalBeijingChina
- Beijing Key Laboratory of Spinal DiseasesBeijingChina
- Engineering Research Center of Bone and Joint Precision MedicineBeijingChina
| |
Collapse
|
20
|
Calcaterra V, Verduci E, Vandoni M, Rossi V, Fiore G, Massini G, Berardo C, Gatti A, Baldassarre P, Bianchi A, Cordaro E, Cavallo C, Cereda C, Bosetti A, Zuccotti G. The Effect of Healthy Lifestyle Strategies on the Management of Insulin Resistance in Children and Adolescents with Obesity: A Narrative Review. Nutrients 2022; 14:4692. [PMID: 36364954 PMCID: PMC9657567 DOI: 10.3390/nu14214692] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 10/29/2022] [Accepted: 11/04/2022] [Indexed: 11/07/2023] Open
Abstract
Childhood obesity is characterized by an increased risk of several metabolic derangements including insulin resistance (IR). The strongest recommendations to prevent obesity and related complications are a balanced and adequate diet and practicing physical activity from early childhood. In this review, we propose to present the effects of healthy lifestyle strategies, including physical exercise and dietary approaches, on the management of IR and related metabolic derangements. All types of exercise (aerobic, resistance and combined training) effectively reduce IR in pediatric patients with obesity; it seems that aerobic and combined training stimulate greater improvements in IR compared to resistance training. Balanced normocaloric or hypocaloric dietary approaches are also valid strategies to address IR; it is not possible to assess the long-term impact of varying macronutrients on cardiometabolic risk. The glycemic index/load evaluation is a useful dietary approach to glucose metabolism control. Similarly, they should adopt the principle of the Mediterranean diet. Randomized studies with longer monitoring are needed to define the benefits of nutritional supplementation on IR. Considering that healthy style acquisition could track to later ages, programs of healthy lifestyle starting with children offer a better preventive strategy to preserve metabolic control and children's health.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, 20154 Milan, Italy
| | - Elvira Verduci
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, 20154 Milan, Italy
- Department of Health Sciences, University of Milano, 20142 Milan, Italy
| | - Matteo Vandoni
- Laboratory of Adapted Motor Activity (LAMA), Department of Public Health, Experimental Medicine and Forensic Science, University of Pavia, 27100 Pavia, Italy
| | - Virginia Rossi
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, 20154 Milan, Italy
| | - Giulia Fiore
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, 20154 Milan, Italy
| | - Giulia Massini
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, 20154 Milan, Italy
| | - Clarissa Berardo
- Department of Biomedical and Clinical Science, University of Milano, 20157 Milan, Italy
| | - Alessandro Gatti
- Laboratory of Adapted Motor Activity (LAMA), Department of Public Health, Experimental Medicine and Forensic Science, University of Pavia, 27100 Pavia, Italy
| | - Paola Baldassarre
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, 20154 Milan, Italy
| | - Alice Bianchi
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, 20154 Milan, Italy
| | - Erika Cordaro
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, 20154 Milan, Italy
| | - Caterina Cavallo
- Laboratory of Adapted Motor Activity (LAMA), Department of Public Health, Experimental Medicine and Forensic Science, University of Pavia, 27100 Pavia, Italy
- LUNEX International University of Health, Exercise and Sports, 50, Avenue du Parc des Sports, 4671 Differdange, Luxembourg
| | - Cristina Cereda
- Neonatal Screening and Metabolic Disorders Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Alessandra Bosetti
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, 20154 Milan, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, 20154 Milan, Italy
- Department of Biomedical and Clinical Science, University of Milano, 20157 Milan, Italy
| |
Collapse
|
21
|
Identifying disease-critical cell types and cellular processes by integrating single-cell RNA-sequencing and human genetics. Nat Genet 2022; 54:1479-1492. [PMID: 36175791 PMCID: PMC9910198 DOI: 10.1038/s41588-022-01187-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/18/2022] [Indexed: 12/13/2022]
Abstract
Genome-wide association studies provide a powerful means of identifying loci and genes contributing to disease, but in many cases, the related cell types/states through which genes confer disease risk remain unknown. Deciphering such relationships is important for identifying pathogenic processes and developing therapeutics. In the present study, we introduce sc-linker, a framework for integrating single-cell RNA-sequencing, epigenomic SNP-to-gene maps and genome-wide association study summary statistics to infer the underlying cell types and processes by which genetic variants influence disease. The inferred disease enrichments recapitulated known biology and highlighted notable cell-disease relationships, including γ-aminobutyric acid-ergic neurons in major depressive disorder, a disease-dependent M-cell program in ulcerative colitis and a disease-specific complement cascade process in multiple sclerosis. In autoimmune disease, both healthy and disease-dependent immune cell-type programs were associated, whereas only disease-dependent epithelial cell programs were prominent, suggesting a role in disease response rather than initiation. Our framework provides a powerful approach for identifying the cell types and cellular processes by which genetic variants influence disease.
Collapse
|
22
|
Regulation of a High-Iron Diet on Lipid Metabolism and Gut Microbiota in Mice. Animals (Basel) 2022; 12:ani12162063. [PMID: 36009656 PMCID: PMC9405328 DOI: 10.3390/ani12162063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/02/2022] Open
Abstract
Iron homeostasis disorder is associated with the imbalance of lipid metabolism, while the specific interaction remains unclear. In the present study, we investigated the effect of a high-iron diet on lipid metabolism in mice. The C57BL/6 mice were fed with a normal diet (WT) or a high-iron diet (WT + Fe) for 12 weeks. We found that mice in the WT + Fe group showed a significant decrease in body weight gain, body fat and lipid accumulation of liver when compared with mice in the WT group. Accordingly, serum total cholesterol and triglyceride levels were both reduced in mice with a high-iron diet. Moreover, mice in the WT + Fe group exhibited a significant decrease in expression of genes regulating adipogenesis and adipocyte differentiation, and a significant increase in expression of fat hydrolysis enzyme genes in both liver and adipose tissues, which was consistent with their dramatic reduction in adipocyte cell size. In addition, a high-iron diet decreased the relative abundance of beneficial bacteria (Akkermansia, Bifidobacterium and Lactobacillus) and increased the relative abundance of pathogenic bacteria (Romboutsia and Erysipelatoclostridium). Thus, our research revealed that a high-iron diet reduced lipid deposition by inhibiting adipogenesis and promoting lipolysis. Altered gut microbial composition induced by a high-iron diet may not play a critical role in regulating lipid metabolism, but might cause unwanted side effects such as intestinal inflammation and damaged villi morphology at the intestinal host–microbe interface. These findings provide new insights into the relationship among iron, lipid metabolism and gut microbiota.
Collapse
|
23
|
Fujimori K. Prostaglandin D<sub>2</sub> and F<sub>2α</sub> as Regulators of Adipogenesis and Obesity. Biol Pharm Bull 2022; 45:985-991. [DOI: 10.1248/bpb.b22-00210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Ko Fujimori
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University
| |
Collapse
|
24
|
Wu J, Du J, Li Z, He W, Wang M, Jin M, Yang L, Liu H. Pentamethylquercetin Regulates Lipid Metabolism by Modulating Skeletal Muscle-Adipose Tissue Crosstalk in Obese Mice. Pharmaceutics 2022; 14:pharmaceutics14061159. [PMID: 35745732 PMCID: PMC9227162 DOI: 10.3390/pharmaceutics14061159] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/21/2022] [Accepted: 05/27/2022] [Indexed: 01/27/2023] Open
Abstract
Irisin is an exercise-induced hormone that regulates lipid metabolism. The present study investigates whether the anti-obesity effect of the natural flavonoid pentamethylquercetin (PMQ) is related to irisin secretion from skeletal muscle in whole animals and cultured cells. Obese mice induced by monosodium glutamate were administered oral PMQ to determine blood irisin level and in vivo parameters of lipid metabolism, and cultured mouse C2C12 myoblasts and 3T3-L1 preadipocytes were employed to investigate the related molecular identities. PMQ increased circulating irisin and decreased bodyweight, insulin, and lipid levels accompanied with increasing brown-like adipocyte formation in obese mice. The brown adipocyte marker uncoupling protein 1 (UCP-1) and other brown-like adipocyte-specific genes and/or markers were increased in mouse white fat tissue, while PMQ treatment reversed the above changes. PMQ also dose-dependently increased the reduced levels of AMP-activated protein kinase (AMPK), peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), and fibronectin type III domain-containing 5 (FNDC5) signal molecules in obese mice. Interestingly, the irisin level was increased in the culture medium of C2C12 cells treated with PMQ, and the conditioned medium stimulated the brown-like transition of 3T3-L1 preadipocytes with the increased expression of PGC-1α, FNDC5, UCP-1, and other brown-like adipocyte-specific genes. The effects of conditioned culture medium were abolished in C2C12 cells with silenced PGC-1α. On the other hand, PMQ-induced upregulation of PGC-1α and FNDC5 expression was reduced by AMPK inhibitor Compound C in C2C12 cells. Our results demonstrate the novel information that PMQ-induced irisin secretion from skeletal muscle involves the improvement of metabolic dysfunction in obese mice via activating the AMPK/PGC-1α/FNDC5 signal pathway, suggesting that PMQ modulates skeletal muscle-adipose tissue crosstalk and may be a promising drug candidate for treating obesity and obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Jianzhao Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.W.); (J.D.); (Z.L.); (W.H.); (M.W.); (M.J.)
| | - Jingxia Du
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.W.); (J.D.); (Z.L.); (W.H.); (M.W.); (M.J.)
| | - Zhi Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.W.); (J.D.); (Z.L.); (W.H.); (M.W.); (M.J.)
| | - Wei He
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.W.); (J.D.); (Z.L.); (W.H.); (M.W.); (M.J.)
| | - Min Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.W.); (J.D.); (Z.L.); (W.H.); (M.W.); (M.J.)
| | - Manwen Jin
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.W.); (J.D.); (Z.L.); (W.H.); (M.W.); (M.J.)
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lei Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Correspondence: (L.Y.); (H.L.)
| | - Hui Liu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.W.); (J.D.); (Z.L.); (W.H.); (M.W.); (M.J.)
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, China
- Correspondence: (L.Y.); (H.L.)
| |
Collapse
|
25
|
Damen MSMA, Alarcon PC, Shah AS, Divanovic S. Greasing the inflammatory pathogenesis of viral pneumonias in diabetes. Obes Rev 2022; 23:e13415. [PMID: 34989117 PMCID: PMC9771603 DOI: 10.1111/obr.13415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/16/2022]
Abstract
Type 2 diabetes (T2D) and obesity are independent risk factors for increased morbidity and mortality associated with influenza and SARS-CoV-2 infection. Skewed cellular metabolism shapes immune cell inflammatory responsiveness and function in obesity, T2D, and infection. However, altered immune cell responsiveness and levels of systemic proinflammatory mediators, partly independent of peripheral immune cell contribution, are linked with SARS-CoV-2-associated disease severity. Despite such knowledge, the role of tissue parenchymal cell-driven inflammatory responses, and specifically those dominantly modified in obesity (e.g., adipocytes), in influenza and SARS-CoV-2 infection pathogenesis remain poorly defined. Whether obesity-dependent skewing of adipocyte cellular metabolism uncovers inflammatory clades and promotes the existence of a 'pathogenic-inflammatory' adipocyte phenotype that amplifies SARS-CoV-2 infection diseases severity in individuals with obesity and individuals with obesity and T2D has not been examined. Here, using the knowledge gained from studies of immune cell responses in obesity, T2D, and infection, we highlight the key knowledge gaps underlying adipocyte cellular functions that may sculpt and grease pathogenic processes associated with influenza and SARS-CoV-2 disease severity in diabetes.
Collapse
Affiliation(s)
- Michelle S M A Damen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Pablo C Alarcon
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, Cincinnati Childrens Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Amy S Shah
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, Cincinnati Childrens Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
26
|
Fujimori K, Uno S, Kuroda K, Matsumoto C, Maehara T. Leukotriene C 4 synthase is a novel PPARγ target gene, and leukotriene C 4 and D 4 activate adipogenesis through cysteinyl LT1 receptors in adipocytes. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119203. [PMID: 34968576 DOI: 10.1016/j.bbamcr.2021.119203] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/07/2021] [Accepted: 12/18/2021] [Indexed: 01/22/2023]
Abstract
Leukotriene (LT) C4 synthase (LTC4S) catalyzes the conversion from LTA4 to LTC4, which is a proinflammatory lipid mediator in asthma and other inflammatory diseases. LTC4 is metabolized to LTD4 and LTE4, all of which are known as cysteinyl (Cys) LTs and exert physiological functions through CysLT receptors. LTC4S is expressed in adipocytes. However, the function of CysLTs and the regulatory mechanism in adipocytes remain unclear. In this study, we investigated the expression of LTC4S and production of CysLTs in murine adipocyte 3T3-L1 cells and their underlying regulatory mechanisms. Expression of LTC4S and production of LTC4 and CysLTs increased during adipogenesis, whereas siRNA-mediated suppression of LTC4S expression repressed adipogenesis by reducing adipogenic gene expression. The CysLT1 receptor, one of the two LTC4 receptors, was expressed in adipocytes. LTC4 and LTD4 increased the intracellular triglyceride levels and adipogenic gene expression, and their enhancement was suppressed by co-treatment with pranlukast, a CysLT1 receptor antagonist. Moreover, the expression profiles of LTC4S gene/protein during adipogenesis resembled those of peroxisome proliferator-activated receptor (PPAR) γ. LTC4S expression was further upregulated by treatment with troglitazone, a PPARγ agonist. Promoter-luciferase and chromatin immunoprecipitation assays showed that PPARγ directly bound to the PPAR response element of the LTC4S gene promoter in adipocytes. These results indicate that the LTC4S gene expression was enhanced by PPARγ, and LTC4 and LTD4 activated adipogenesis through CysLT1 receptors in 3T3-L1 cells. Thus, LTC4S and CysLT1 receptors are novel potential targets for the treatment of obesity.
Collapse
Affiliation(s)
- Ko Fujimori
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| | - Saki Uno
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan
| | - Kyohei Kuroda
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan
| | - Chihiro Matsumoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan
| | - Toko Maehara
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan
| |
Collapse
|
27
|
Patil AD, Aphale PS, Sharma DB, Bhonde RR. Can homeopathic medicine Chelidonium majus serve a dual role of an anti-obesity and anti-diabetic agent? Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2021.110749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
28
|
Vitale E. A Chronic Inflammatory Inductive Condition in the Nursing Profession: A Scoping Review. Endocr Metab Immune Disord Drug Targets 2022; 22:1235-1244. [PMID: 35578870 DOI: 10.2174/1871530322666220516163936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Literature focuses on the well-being of patients and little on the wellbeing of nurses who, in turn, should be educated about well-being. On the other hand, the latter often work under serious pressure with inadequate resources and shift organizations, exposing them to health risk factors. OBJECTIVE To highlight which relationships exist between diet, physical activity, body mass index (BMI) and mental health in nursing. METHODS The author searched the Medline (PubMed) database for medical subheadings terms and free full text referred to "Diet," "Mental Health," "Physical Activity," and "Nurses" before 31st December 2020. RESULTS A total of 11 studies were included in this perspective review, which better defined the relationships between diet, physical activity, mental health and the nursing profession. CONCLUSION From the studies present in the literature, it emerges that the nursing profession has an inherent imbalance in the regular circadian activity of human nature, as nursing assistance is provided 24 hours a day and during the night. Numerous studies have highlighted significant differences between mental and eating disorder levels among nurses who work at night versus those who only work during the day. Therefore, it is possible to speak of the nursing profession as an "inductive inflammatory-chronic" activity based on the extensive scientific evidence provided by the literature.
Collapse
Affiliation(s)
- Elsa Vitale
- Centre of Mental Health Modugno, Local Health Authority Bari, Bari, Italy
| |
Collapse
|
29
|
|
30
|
McColloch A, Liu H, Cho M. Reversal of stem cell‐derived hypertrophic adipocytes mediated by photobiomodulation (1064 nm). TRANSLATIONAL BIOPHOTONICS 2021. [DOI: 10.1002/tbio.202100006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Andrew McColloch
- Department of Bioengineering The University of Texas at Arlington Arlington Texas USA
| | - Hanli Liu
- Department of Bioengineering The University of Texas at Arlington Arlington Texas USA
| | - Michael Cho
- Department of Bioengineering The University of Texas at Arlington Arlington Texas USA
| |
Collapse
|
31
|
Jagadeesh KA, Dey KK, Montoro DT, Mohan R, Gazal S, Engreitz JM, Xavier RJ, Price AL, Regev A. Identifying disease-critical cell types and cellular processes across the human body by integration of single-cell profiles and human genetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.03.19.436212. [PMID: 34845454 PMCID: PMC8629197 DOI: 10.1101/2021.03.19.436212] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Genome-wide association studies (GWAS) provide a powerful means to identify loci and genes contributing to disease, but in many cases the related cell types/states through which genes confer disease risk remain unknown. Deciphering such relationships is important for identifying pathogenic processes and developing therapeutics. Here, we introduce sc-linker, a framework for integrating single-cell RNA-seq (scRNA-seq), epigenomic maps and GWAS summary statistics to infer the underlying cell types and processes by which genetic variants influence disease. We analyzed 1.6 million scRNA-seq profiles from 209 individuals spanning 11 tissue types and 6 disease conditions, and constructed gene programs capturing cell types, disease progression, and cellular processes both within and across cell types. We evaluated these gene programs for disease enrichment by transforming them to SNP annotations with tissue-specific epigenomic maps and computing enrichment scores across 60 diseases and complex traits (average N= 297K). Cell type, disease progression, and cellular process programs captured distinct heritability signals even within the same cell type, as we show in multiple complex diseases that affect the brain (Alzheimer’s disease, multiple sclerosis), colon (ulcerative colitis) and lung (asthma, idiopathic pulmonary fibrosis, severe COVID-19). The inferred disease enrichments recapitulated known biology and highlighted novel cell-disease relationships, including GABAergic neurons in major depressive disorder (MDD), a disease progression M cell program in ulcerative colitis, and a disease-specific complement cascade process in multiple sclerosis. In autoimmune disease, both healthy and disease progression immune cell type programs were associated, whereas for epithelial cells, disease progression programs were most prominent, perhaps suggesting a role in disease progression over initiation. Our framework provides a powerful approach for identifying the cell types and cellular processes by which genetic variants influence disease.
Collapse
|
32
|
Yamamoto M, Nagasawa Y, Fujimori K. Glycyrrhizic acid suppresses early stage of adipogenesis through repression of MEK/ERK-mediated C/EBPβ and C/EBPδ expression in 3T3-L1 cells. Chem Biol Interact 2021; 346:109595. [PMID: 34302803 DOI: 10.1016/j.cbi.2021.109595] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022]
Abstract
Glycyrrhizic acid (GA), a major constituent of the root of licorice (Glycyrrhiza glabra), and has various biological activities, including anti-obesity property. However, the molecular mechanism of anti-adipogenic effect of GA is still unclear. In this study, we investigated the anti-adipogenic effects of GA in mouse adipocytic 3T3-L1 cells and elucidated its underlying molecular mechanism. GA decreased the intracellular triglyceride level. The expression levels of the adipogenic and lipogenic genes were lowered by treatment with GA in a concertation-dependent manner. In contrast, GA did not affect the lipolytic gene expression and the released glycerol level. GA suppressed the early stage of adipogenesis when it was added for 0-3 h after initiation of adipogenesis. Moreover, GA reduced the mRNA levels of CCAAT/enhancer binding protein (C/EBP) β and C/EBPδ, both of which activate the early stage of adipogenesis. Furthermore, GA decreased phosphorylation of extracellular signal-regulated kinase [ERK: p44/42 mitogen-activated protein kinase (MAPK)] in the early stage of adipogenesis. In addition, a MAPK kinase (MEK) inhibitor, PD98059 reduced the C/EBPβ and C/EBPδ gene expression. These results indicate that GA suppressed the early stage of adipogenesis through repressing the MEK/ERK-mediated C/EBPβ and C/EBPδ expression in 3T3-L1 cells. Thus, GA has an anti-adipogenic ability and a possible agent for treatment of obesity.
Collapse
Affiliation(s)
- Masayuki Yamamoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Yasuna Nagasawa
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Ko Fujimori
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan.
| |
Collapse
|
33
|
Ortiz-Huidobro RI, Velasco M, Larqué C, Escalona R, Hiriart M. Molecular Insulin Actions Are Sexually Dimorphic in Lipid Metabolism. Front Endocrinol (Lausanne) 2021; 12:690484. [PMID: 34220716 PMCID: PMC8251559 DOI: 10.3389/fendo.2021.690484] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/31/2021] [Indexed: 12/14/2022] Open
Abstract
The increment in energy-dense food and low physical activity has contributed to the current obesity pandemic, which is more prevalent in women than in men. Insulin is an anabolic hormone that regulates the metabolism of lipids, carbohydrates, and proteins in adipose tissue, liver, and skeletal muscle. During obesity, nutrient storage capacity is dysregulated due to a reduced insulin action on its target organs, producing insulin resistance, an early marker of metabolic dysfunction. Insulin resistance in adipose tissue is central in metabolic diseases due to the critical role that this tissue plays in energy homeostasis. We focused on sexual dimorphism on the molecular mechanisms of insulin actions and their relationship with the physiology and pathophysiology of adipose tissue. Until recently, most of the physiological and pharmacological studies were done in males without considering sexual dimorphism, which is relevant. There is ample clinical and epidemiological evidence of its contribution to the establishment and progression of metabolic diseases. Sexual dimorphism is a critical and often overlooked factor that should be considered in design of sex-targeted therapeutic strategies and public health policies to address obesity and diabetes.
Collapse
Affiliation(s)
- Rosa Isela Ortiz-Huidobro
- Neurosciences Division, Department of Cognitive Neuroscience, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Myrian Velasco
- Neurosciences Division, Department of Cognitive Neuroscience, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Carlos Larqué
- Department of Embryology and Genetics, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Rene Escalona
- Department of Embryology and Genetics, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Marcia Hiriart
- Neurosciences Division, Department of Cognitive Neuroscience, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- *Correspondence: Marcia Hiriart,
| |
Collapse
|
34
|
Sylus AM, Nandeesha H, Chitra T. Matrix metalloproteinase-9 increases and Interleukin-10 reduces with increase in body mass index in polycystic ovary syndrome: A cross-sectional study. Int J Reprod Biomed 2020; 18:605-610. [PMID: 32923927 PMCID: PMC7457150 DOI: 10.18502/ijrm.v13i8.7502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 10/21/2019] [Accepted: 02/22/2020] [Indexed: 12/21/2022] Open
Abstract
Background Obesity, inflammation and alterations in matrix metalloproteinase-9 (MMP-9) and nitric oxide (NO) levels are involved in the development of polycystic ovary syndrome (PCOS). Objective To investigate the relationship of MMP-9, NO and interleukin-10 (IL-10) with the increase in body mass index (BMI) in women with PCOS. Materials and Methods Sixty two infertile PCOS women were included in the study. Serum levels of NO, IL-10 and MMP-9 were assessed in the women with increase in BMI. Results MMP-9 was significantly increased (p = 0.029) and IL-10 (p = 0.015) was significantly reduced in obese PCOS subjects compared to those with lesser BMI. MMP-9 levels positively correlated with the duration of infertility (r = 0.253, p = 0.047) and negatively correlated with NO levels (r = - 0.259, p = 0.042). A significant negative correlation between the interleukin-10 levels and the BMI (r = - 0.272, p = 0.033) was also found in the PCOS subjects. Conclusion MMP-9 levels are increased in obese PCOS women and it is associated with NO levels and the duration of infertility.
Collapse
Affiliation(s)
- Angel Mercy Sylus
- Department of Biochemistry, Jawaharlal Institute of Post Graduate Medical Education and Research, Puducherry, India
| | - Hanumanthappa Nandeesha
- Department of Biochemistry, Jawaharlal Institute of Post Graduate Medical Education and Research, Puducherry, India
| | - Thiagaraju Chitra
- Obstetrics and Gynecology, Jawaharlal Institute of Post Graduate Medical Education and Research, Puducherry, India
| |
Collapse
|
35
|
Maheshwari RA, Parmar GR, Hinsu D, Seth AK, Balaraman R. Novel therapeutic intervention of coenzyme Q10 and its combination with pioglitazone on the mRNA expression level of adipocytokines in diabetic rats. Life Sci 2020; 258:118155. [PMID: 32735887 DOI: 10.1016/j.lfs.2020.118155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/15/2020] [Accepted: 07/23/2020] [Indexed: 01/11/2023]
Abstract
AIMS Aim of the present study was to investigate the effect of co-administration coenzyme Q10 and pioglitazone on the mRNA expression of adipocytokines in white adipose tissues of chemically induced type 2 diabetes mellitus in rats. MAIN METHODS Diabetes was induced by administration of streptozotocin (65 mg/kg, i.p.), followed by nicotinamide (110 mg/kg, i.p.) 15 min later. The diabetic rats were treated coenzyme Q10 (Q10, 10 mg/kg, p.o.) or pioglitazone (PIO, 20 mg/kg, p.o.) alone and their combination for four weeks. Biochemical parameters like FBS level, insulin and HbA1c along with tissue levels of MDA, SOD, CAT and GSH were estimated. The mRNA levels of ADIPOQ, RBP4, RETN, IL-6 and TNF-α in White Adipose Tissue (WAT) were measured. KEY FINDINGS Treatment with Q10 + PIO showed a significant reduction in the levels of FBS, HbA1c and a significant increase in insulin levels as compared to normal control group. Additionally, there was a significant change in the levels of biomarkers of oxidative stress after treatment with Q10 + PIO as compared to streptozotocin-nicotinamide group. Treatment with Q10 + PIO also significantly altered the mRNA expression of ADIPOQ, RETN, IL-6 and TNF-α when compared to monotherapy. However, mRNA expression of RBP4 did not alter in Q10 + PIO treated animal as compared to Q10 or PIO alone. SIGNIFICANCE It is concluded that co-administration of Q10 and PIO has been shown the better therapeutic effect on the mRNA expression of adipocytokines and oxidative stress parameters as compared to either Q10 or PIO.
Collapse
Affiliation(s)
- Rajesh A Maheshwari
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, At Post Piparia, Taluka: Waghodia, Dist: Vadodara, Vadodara-391760, Gujarat, India
| | - Ghanshyam R Parmar
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, At Post Piparia, Taluka: Waghodia, Dist: Vadodara, Vadodara-391760, Gujarat, India.
| | - Denish Hinsu
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, At Post Piparia, Taluka: Waghodia, Dist: Vadodara, Vadodara-391760, Gujarat, India
| | - Avinash K Seth
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, At Post Piparia, Taluka: Waghodia, Dist: Vadodara, Vadodara-391760, Gujarat, India
| | - Ramachandran Balaraman
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, At Post Piparia, Taluka: Waghodia, Dist: Vadodara, Vadodara-391760, Gujarat, India
| |
Collapse
|
36
|
Promotion of lipogenesis by PPARγ-activated FXR expression in adipocytes. Biochem Biophys Res Commun 2020; 527:49-55. [DOI: 10.1016/j.bbrc.2020.04.075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 12/11/2022]
|
37
|
Antiadipogenic Effects of Mixtures of Cornus officinalis and Ribes fasciculatum Extracts on 3T3-L1 Preadipocytes and High-Fat Diet-Induced Mice. Molecules 2020; 25:molecules25102350. [PMID: 32443487 PMCID: PMC7287608 DOI: 10.3390/molecules25102350] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
Medicinal plants have been used worldwide as primary alternative healthcare supplements. Cornus officinalis (CO) and Ribes fasciculatum (RF) are traditional medicinal plants applied in East Asia to treat human diseases such as hepatitis, osteoporosis, oxidative stress and allergy. The aim of this study was to examine the anti-obesity effect of CO and RF on preadipocyte 3T3-L1 cells in vitro and high-fat diet (HFD)-induced obesity mice in vivo. Combination treatment of CO and RF in differentiated 3T3-L1 cells inhibited adipocyte differentiation through downregulation of adipogenesis-associated genes such as CCAAT/enhancer-binding protein alpha (Cebpa), fatty acid binding protein 4 (Fabp4), peroxisome proliferator-activated receptor gamma (Pparg) and sterol regulatory element binding protein (Srebp1). In vivo animal models showed that a mixture of CO and RF inhibited HFD-induced weight gain, resulting in decreased abdominal visceral fat tissues and fatty hepatocyte deposition. In addition, CO+RF treatment decreased HFD-induced adipogenesis-associated genes in abdominal white fat tissue. These results suggest that administration of a CO and RF mixture prevented adipocyte differentiation and lipid accumulation in preadipocyte cells and HFD-induced body weight in obesity mice. Therefore, combined therapy of CO and RF may be a protective therapeutic agent against obesity.
Collapse
|
38
|
Abdel Aziz SM, Ahmed OM, Abd EL-Twab SM, Al-Muzafar HM, Amin KA, Abdel-Gabbar M. Antihyperglycemic Effects and Mode of Actions of Musa paradisiaca Leaf and Fruit Peel Hydroethanolic Extracts in Nicotinamide/Streptozotocin-Induced Diabetic Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:9276343. [PMID: 32047529 PMCID: PMC7007756 DOI: 10.1155/2020/9276343] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
The present study aimed to evaluate the antihyperglycemic effects of Musa paradisiaca (M. paradisiaca) leaf and fruit peel hydroethanolic extracts and to suggest their probable mode of actions in nicotinamide (NA)/streptozotocin (STZ)-induced diabetic rats. The leaf and fruit peel hydroethanolic extracts were analyzed by GC-MS that indicated the presence of phytol, octadecatrienoic acid, hexadecanoic acid, and octadecadienoic acid as major components in the leaf extract and vitamin E, octadecenamide, β-sitosterol, and stigmasterol as major phytochemicals in the fruit peel extract. Diabetes mellitus was induced by a single intraperitoneal injection of STZ (60 mg/kg body weight) dissolved in citrate buffer (pH 4.5), 15 minutes after intraperitoneal injection of NA (120 mg/kg body weight). The NA/STZ-induced diabetic rats were, respectively, treated with M. paradisiaca leaf and fruit peel hydroethanolic extracts at a dose of 100 mg/kg body weight/day by oral administration for 28 days. The treatment of NA/STZ-induced diabetic rats with leaf and fruit peel extracts significantly improved the impaired oral glucose tolerance and significantly increased the lowered serum insulin and C-peptide levels. The HOMA-IR (as the index of insulin resistance) and QUICKI (as a marker for insulin sensitivity), as well as HOMA-β cell function were significantly alleviated as a result of treatment of diabetic rats with leaf and fruit peel extracts. In association, the elevated serum-free fatty acids, TNF-α, and IL-6 levels were significantly decreased. In addition, the suppressed adipose tissue PPARγ, GLUT4, adiponectin, and insulin receptor β-subunit mRNA expressions were upregulated while the elevated adipose tissue resistin expression was downregulated in diabetic rats as a result of treatment with the leaf and peel extract. Based on these results, it can be concluded that M. paradisiaca leaf and fruit peel hydroethanolic extracts have antihyperglycemic effects which may be mediated via their insulinotropic and insulin-sensitizing effects.
Collapse
Affiliation(s)
- Sarah M. Abdel Aziz
- Biochemistry Division, Chemistry Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Osama M. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Sanaa M. Abd EL-Twab
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Hessah Mohammed Al-Muzafar
- Chemistry Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box. 1982, Dammam 31441, Saudi Arabia
| | - Kamal Adel Amin
- Chemistry Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box. 1982, Dammam 31441, Saudi Arabia
- Basic & Applied Scientific Research Center, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Mohamed Abdel-Gabbar
- Biochemistry Division, Chemistry Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| |
Collapse
|
39
|
Basen-Engquist K, Brown P, Coletta AM, Savage M, Maresso KC, Hawk E. Lifestyle and Cancer Prevention. ABELOFF'S CLINICAL ONCOLOGY 2020:337-374.e12. [DOI: 10.1016/b978-0-323-47674-4.00022-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
40
|
WDR76 mediates obesity and hepatic steatosis via HRas destabilization. Sci Rep 2019; 9:19676. [PMID: 31873167 PMCID: PMC6927951 DOI: 10.1038/s41598-019-56211-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 12/03/2019] [Indexed: 12/22/2022] Open
Abstract
Ras/MAPK (mitogen active protein kinase) signaling plays contradictory roles in adipocyte differentiation and is tightly regulated during adipogenesis. However, mechanisms regulating adipocyte differentiation involving Ras protein stability regulation are unknown. Here, we show that WD40 repeat protein 76 (WDR76), a novel Ras regulating E3 linker protein, controls 3T3-L1 adipocyte differentiation through HRas stability regulation. The roles of WDR76 in obesity and metabolic regulation were characterized using a high-fat diet (HFD)-induced obesity model using Wdr76-/- mice and liver-specific Wdr76 transgenic mice (Wdr76Li-TG). Wdr76-/- mice are resistant to HFD-induced obesity, insulin resistance and hyperlipidemia with an increment of HRas levels. In contrast, Wdr76Li-TG mice showed increased HFD-induced obesity, insulin resistance with reduced HRas levels. Our findings suggest that WDR76 controls HFD-induced obesity and hepatic steatosis via HRas destabilization. These data provide insights into the links between WDR76, HRas, and obesity.
Collapse
|
41
|
Sun Y, Wang Y, Song P, Wang H, Xu N, Wang Y, Zhang Z, Yue P, Gao X. Anti-obesity effects of instant fermented teas in vitro and in mice with high-fat-diet-induced obesity. Food Funct 2019; 10:3502-3513. [PMID: 31143917 DOI: 10.1039/c9fo00162j] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity is a chronic metabolic disorder that is associated with higher risks of developing diabetes and cardiovascular disease. Chinese dark tea is a fermented beverage with many biological effects and could be considered for the management of obesity. This study is aimed to assess the possible anti-obesity properties of instant dark tea (IDT) and instant pu-erh tea (PET) in high fat diet (HFD)-fed mice. Male C57BL/6 mice were divided into 5 groups. They received low-fat diet (LFD), HFD, HFD supplemented with drinking IDT infusion (5 mg mL-1), PET infusion (5 mg mL-1) or water for 8 weeks. The results showed IDT exhibited better inhibitory effect than PET on body weight gain and visceral fat weights. IDT also improved the serum high-density lipoprotein cholesterol (HDL-C) level, but decreased the low-density lipoprotein cholesterol (LDL-C) and leptin levels more effectively than PET. Both IDT and PET lowered the levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in the plasma and significantly increased the ratio of albumin to globin (A/G) in the serum compared to the control group. IDT treatment reduced the malondialdehyde (MDA) level in the liver. Histomorphology evidenced that the liver tissue architecture was well preserved by IDT administration. Moreover, IDT regulated the expression of obesity-related genes more effectively than PET. Overall, the present findings have provided the proof of concept that dietary IDT could provide a safer and cost-effective option for people with HFD-induced obesity.
Collapse
Affiliation(s)
- Yue Sun
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Shaaban Z, Khoradmehr A, Amiri-Yekta A, Jafarzadeh Shirazi MR, Tamadon A. Pathophysiologic mechanisms of obesity- and chronic inflammation-related genes in etiology of polycystic ovary syndrome. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:1378-1386. [PMID: 32133054 PMCID: PMC7043875 DOI: 10.22038/ijbms.2019.14029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 06/25/2019] [Indexed: 01/02/2023]
Abstract
OBJECTIVES One of the common heterogeneous reproductive disorders in women of childbearing age is polycystic ovary syndrome (PCOS). It is characterized by lack of fertility due to anovulatory cycles, hyperandrogenemia, polycystic ovaries, hyperinsulinemia, and obesity. Both reproductive anomalies and metabolic disorders are involved in PCOS pathology. Although the role of increased levels of androgens in initiation of PCOS is almost proven, mechanisms of PCOS pathophysiology are not clear. Here we discuss roles of altered metabolic conditions, obesity, and chronic inflammation in PCOS pathophysiology. MATERIALS AND METHODS In this review, we attempted to identify genes related to obesity and chronic inflammation aspects of PCOS and their physiological functions to explain the pathways that are regulated by these genes and can be a prominent function in PCOS predisposition. For this purpose, published articles and reviews dealing with genetic evaluation of PCOS in women in peer-reviewed journals in PubMed and Google Scholar databases were included in this review. RESULTS Obesity and chronic inflammation are not prominent diagnostic features of PCOS, but they play an important role in exacerbating metabolic and hyperandrogenic states. ADIPOQ, FTO TGFβ, and DENND1A as the main obesity- and chronic inflammation-related genes have roles in PCOS pathophysiology. CONCLUSION It seems that genes related to obesity pathology in genomic research association, are related to metabolic aspects and body mass index in PCOS patients. Genomes have roles in chronic inflammation, followed by obesity, in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Zahra Shaaban
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran
| | - Arezoo Khoradmehr
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Amir Amiri-Yekta
- Reproductive Biomedicine Research Center, Royan Institute, Tehran, Iran
| | | | - Amin Tamadon
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
43
|
Sun Y, Li L, Wu J, Gong B, Liu H. Germacrone cooperates with dexmedetomidine to alleviate high-fat diet-induced type 2 diabetes mellitus via upregulating AMPKα1 expression. Exp Ther Med 2019; 18:3514-3524. [PMID: 31602228 PMCID: PMC6777304 DOI: 10.3892/etm.2019.7990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 06/27/2019] [Indexed: 12/11/2022] Open
Abstract
The aim of the present study was to investigate the effects of germacrone (GM) and dexmedetomidine (DEX) in treating type 2 diabetes mellitus (T2DM). A high-fat diet (HFD)-induced T2DM rat model was established. The experimental rats were divided into the control group, HFD group, GM treatment group, DEX treatment group and GM + DEX treatment group. In addition, adenosine monophosphate-activated protein kinase (AMPK) inhibitor compound C (CC) was used to inhibit AMPKα1 expression. All rats received their respective treatment daily for 21 days. Blood glucose and lipid levels, apoptosis of hepatic cells, and levels of inflammatory factors and oxidative stress indicators in serum samples were evaluated. Protein expression of AMPKα1 and its downstream targets were also investigated. Results demonstrated that blood glucose concentration, blood lipid indicators (endothelin, total cholesterol, triglyceride and low density lipoprotein cholesterol), cell apoptosis in liver tissues, total oxidant status, malondialdehyde, interleukin (IL)-6, tumor necrosis factor-α (TNF-α) and IL-1β levels in serum were increased in the high-fat group compared to the control but decreased following GM and/or DEX treatment. By contrast, high-density lipoprotein cholesterol and antioxidative stress indicator superoxide dismutase (SOD) were decreased in the high-fat group but increased following GM and/or DEX treatment. Protein expression of AMPKα1 and the catabolic genes carnitine palmitoyltransferase-1, peroxisome proliferator-activated receptor-α and acyl coenzyme A were decreased whilst anabolic genes, including sterol regulatory element binding protein-1c, fatty acid synthase and diacylglycerol acyltransferase-2, were increased in the HFD group. These effects were attenuated by GM and/or DEX treatment. AMPKα1 inhibition resulted in decreased SOD and increased cell apoptosis in liver tissues as well as increased IL-6, TNF-α and IL-1β levels compared with the HFD group. However, these effects were abolished following treatment with CC, GM and DEX together. Taken together these results indicated that GM worked synergistically with DEX to attenuate symptoms of high-fat-induced T2DM, with the effect potentially involving an increase in AMPKα1 expression.
Collapse
Affiliation(s)
- Yang Sun
- Department of Anesthesia, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Lanlan Li
- Department of Anesthesia, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Jun Wu
- Department of Anesthesia, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Bing Gong
- Department of Anesthesia, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Haiyan Liu
- Department of Anesthesia, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| |
Collapse
|
44
|
Matsumoto C, Maehara T, Tanaka R, Fujimori K. Limonoid 7-Deacetoxy-7-oxogedunin from Andiroba, Carapa guianensis, Meliaceae, Decreased Body Weight Gain, Improved Insulin Sensitivity, and Activated Brown Adipose Tissue in High-Fat-Diet-Fed Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:10107-10115. [PMID: 31434473 DOI: 10.1021/acs.jafc.9b04362] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We examined the antiobesity effect of a limonoid 7-deacetoxy-7-oxogedunin, named CG-1, purified from the seeds of Carapa guianensis, Meliaceae, known as andiroba in high-fat-diet (HFD)-fed mice. C57BL/6 mice were fed a low-fat diet or an HFD and orally administered CG-1 (20 mg/kg) for 7 weeks. CG-1 lowered the body weight gain and improved the serum triglyceride level and insulin sensitivity in HFD-fed mice. The expression level of the adipogenesis-related genes was lowered by CG-1 in the visceral white adipose tissue (vWAT). The mRNA expression level of the macrophage-related genes decreased in vWAT following the administration of CG-1 to HFD-fed mice. It is noteworthy that CG-1 activated the brown adipose tissue (BAT) with enhanced expression of uncoupling protein 1 and increased the rectal temperature in HFD-fed mice. These results indicate that the limonoid CG-1 decreased body weight gain and ameliorated hypertriglyceridemia and insulin resistance with the activation of BAT in HFD-fed mice.
Collapse
|
45
|
Lei X, Wong GW. C1q/TNF-related protein 2 (CTRP2) deletion promotes adipose tissue lipolysis and hepatic triglyceride secretion. J Biol Chem 2019; 294:15638-15649. [PMID: 31439668 DOI: 10.1074/jbc.ra119.009230] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/16/2019] [Indexed: 11/06/2022] Open
Abstract
The highly conserved C1q/TNF-related protein (CTRP) family of secreted hormones has emerged as important regulators of insulin action and of sugar and fat metabolisms. Among these, the specific biological function of CTRP2 remains elusive. Here, we show that the expression of human CTRP2 is positively correlated with body mass index (BMI) and is up-regulated in obesity. We used a knockout (KO) mouse model to determine CTRP2 function and found that Ctrp2-KO mice have significantly elevated metabolic rates and energy expenditure leading to lower body weights and lower adiposity. CTRP2 deficiency up-regulated the expression of lipolytic enzymes and protein kinase A signaling, resulting in enhanced adipose tissue lipolysis. In cultured adipocytes, CTRP2 treatment suppressed triglyceride (TG) hydrolysis, and its deficiency enhanced agonist-induced lipolysis in vivo CTRP2-deficient mice also had altered hepatic and plasma lipid profiles. Liver size and hepatic TG content were significantly reduced, but plasma TG was elevated in KO mice. Both plasma and hepatic cholesterol levels, however, were reduced in KO mice. Loss of CTRP2 also enhanced hepatic TG secretion and contributed to impaired plasma lipid clearance following an oral lipid gavage. Liver metabolomic analysis revealed significant changes in diacylglycerols and phospholipids, suggesting that increased membrane remodeling may underlie the altered hepatic TG secretion we observed. Our results provide the first in vivo evidence that CTRP2 regulates lipid metabolism in adipose tissue and liver.
Collapse
Affiliation(s)
- Xia Lei
- Department of Physiology and Center for Metabolism and Obesity Research, School of Medicine, The Johns Hopkins University, Baltimore, Maryland 21205
| | - G William Wong
- Department of Physiology and Center for Metabolism and Obesity Research, School of Medicine, The Johns Hopkins University, Baltimore, Maryland 21205
| |
Collapse
|
46
|
Dual-Energy X-Ray Absorptiometry Compared to Computed Tomography for Visceral Adiposity Assessment Among Gastrointestinal and Pancreatic Cancer Survivors. Sci Rep 2019; 9:11500. [PMID: 31395928 PMCID: PMC6687706 DOI: 10.1038/s41598-019-48027-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 07/23/2019] [Indexed: 01/07/2023] Open
Abstract
Dual-energy x-ray absorptiometry (DXA) for visceral adipose tissue (VAT) assessment is used as an alternative to computed tomography (CT) for research purposes in apparently healthy and clinical populations. It is unknown whether DXA is comparable to CT among cancer survivors, especially in cases where VAT assessment may be affected by treatment history and side effects and become more challenging to assess, such as a history of surgical gastrointestinal resection and/or ascites. The purpose of this study was to determine the level of agreement between DXA and CT when assessing VAT area and volume among cancer survivors. One hundred Gastrointestinal and pancreatic cancer survivors underwent abdominal and pelvis CT and whole-body DXA within 48 hours. Bland-Altman analysis revealed that in women and men, DXA VAT-area estimates were larger and smaller, respectively, and was consistently smaller in estimates for VAT-volume. Correlations from linear regression analysis revealed statistically significant positive correlations between measurement methods. Overall, while DXA VAT estimates are highly correlated with CT VAT estimates, DXA estimates show substantial bias which indicates the two methods are not interchangeable in this population. Further research is warranted with a larger, more homogeneous sample to develop better estimates of the bias.
Collapse
|
47
|
Cao Q, Zhang J, Yu Q, Wang J, Dai M, Zhang Y, Luo Q, Bao M. Carotid baroreceptor stimulation in obese rats affects white and brown adipose tissues differently in metabolic protection. J Lipid Res 2019; 60:1212-1224. [PMID: 31126973 DOI: 10.1194/jlr.m091256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 05/23/2019] [Indexed: 11/20/2022] Open
Abstract
The sympathetic nervous system (SNS) regulates the functions of white adipose tissue (WAT) and brown adipose tissue (BAT) tightly. Carotid baroreceptor stimulation (CBS) efficiently inhibits SNS activation. We hypothesized that CBS would protect against obesity. We administered CBS to obese rats and measured sympathetic and AMP-activated protein kinase (AMPK)/ PPAR pathway responses as well as changes in perirenal WAT (PWAT), epididymal WAT (EWAT), and interscapular BAT (IBAT). CBS alleviated obesity-related metabolic changes, improving insulin resistance; reducing adipocyte hypertrophy, body weight, and adipose tissue weights; and decreasing norepinephrine but increasing acetylcholine in plasma, PWAT, EWAT, and IBAT. CBS also downregulated fatty acid translocase (CD36), fatty acid transport protein (FATP), phosphorylated and total hormone sensitive lipase, phosphorylated and total protein kinase A, and PPARγ in obese rats. Simultaneously, CBS upregulated phosphorylated adipose triglyceride lipase, phosphorylated and total AMPK, and PPARα in PWAT, EWAT, and IBAT. However, BAT and WAT responses differed; although many responses were more sensitive in IBAT, responses of CD36, FATP, and PPARγ were more sensitive in PWAT and EWAT. Overall, CBS decreased chronically activated SNS and ameliorated obesity-related metabolic disorders by regulating the AMPK/PPARα/γ pathway.
Collapse
Affiliation(s)
- Quan Cao
- Department of Cardiology, Renmin Hospital of Wuhan University.,Cardiovascular Research Institute Wuhan University.,Hubei Key Laboratory of Cardiology Wuhan 430060, China
| | - Junxia Zhang
- Department of Endocrinology, Wuhan General Hospital of the Chinese People's Liberation Army, Wuhan 430060, China
| | - Qiao Yu
- Department of Cardiology, Renmin Hospital of Wuhan University.,Cardiovascular Research Institute Wuhan University.,Hubei Key Laboratory of Cardiology Wuhan 430060, China
| | - Jing Wang
- Department of Cardiology, Renmin Hospital of Wuhan University.,Cardiovascular Research Institute Wuhan University.,Hubei Key Laboratory of Cardiology Wuhan 430060, China
| | - Mingyan Dai
- Department of Cardiology, Renmin Hospital of Wuhan University.,Cardiovascular Research Institute Wuhan University
| | - Yijie Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University.,Cardiovascular Research Institute Wuhan University.,Hubei Key Laboratory of Cardiology Wuhan 430060, China
| | - Qiang Luo
- Department of Cardiology, Renmin Hospital of Wuhan University.,Cardiovascular Research Institute Wuhan University.,Hubei Key Laboratory of Cardiology Wuhan 430060, China
| | - Mingwei Bao
- Department of Cardiology, Renmin Hospital of Wuhan University .,Cardiovascular Research Institute Wuhan University.,Hubei Key Laboratory of Cardiology Wuhan 430060, China
| |
Collapse
|
48
|
Bond ST, Kim J, Calkin AC, Drew BG. The Antioxidant Moiety of MitoQ Imparts Minimal Metabolic Effects in Adipose Tissue of High Fat Fed Mice. Front Physiol 2019; 10:543. [PMID: 31139092 PMCID: PMC6517842 DOI: 10.3389/fphys.2019.00543] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction is associated with a diverse array of diseases ranging from dystrophy and heart failure to obesity and hepatosteatosis. One of the major biochemical consequences of impaired mitochondrial function is an accumulation of mitochondrial superoxide, or reactive oxygen species (ROS). Excessive ROS can be detrimental to cellular health and is proposed to underpin many mitochondrial diseases. Accordingly, much research has been committed to understanding ways to therapeutically prevent and reduce ROS accumulation. In white adipose tissue (WAT), ROS is associated with obesity and its subsequent complications, and thus reducing mitochondrial ROS may represent a novel strategy for treating obesity related disorders. One therapeutic approach employed to reduce ROS abundance is the mitochondrial-targeted coenzyme Q (MitoQ), which enables mitochondrial specific delivery of a CoQ10 antioxidant via its triphenylphosphonium bromide (TPP+) cation. Indeed, MitoQ has been successfully shown to accumulate at the outer mitochondrial membrane and prevent ROS accumulation in several tissues in vivo; however, the specific effects of MitoQ on adipose tissue metabolism in vivo have not been studied. Here we demonstrate that mice fed high-fat diet with concomitant administration of MitoQ, exhibit minimal metabolic benefit in adipose tissue. We also demonstrate that both MitoQ and its control agent dTPP+ had significant and equivalent effects on whole-body metabolism, suggesting that the dTPP+ cation rather than the antioxidant moiety, was responsible for these changes. These findings have important implications for future studies using MitoQ and other TPP+ compounds.
Collapse
Affiliation(s)
- Simon T Bond
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jisu Kim
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Anna C Calkin
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
49
|
Matsumoto C, Koike A, Tanaka R, Fujimori K. A Limonoid, 7-Deacetoxy-7-Oxogedunin (CG-1) from Andiroba ( Carapa guianensis, Meliaceae) Lowers the Accumulation of Intracellular Lipids in Adipocytes via Suppression of IRS-1/Akt-Mediated Glucose Uptake and a Decrease in GLUT4 Expression. Molecules 2019; 24:molecules24091668. [PMID: 31035366 PMCID: PMC6540142 DOI: 10.3390/molecules24091668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 04/25/2019] [Accepted: 04/26/2019] [Indexed: 01/28/2023] Open
Abstract
Limonoids are phytochemicals with a variety of biological properties. In the present study, we elucidated the molecular mechanism of suppression of adipogenesis in adipocytes by a limonoid, 7-deacetoxy-7-oxogedunin (CG-1) from Carapa guianensis (Meliaceae), known as andiroba. CG-1 reduced the accumulation of intracellular triglycerides in a concentration-dependent manner. The expression levels of the adipogenic, lipogenic, and lipolytic genes were decreased by CG-1 treatment, whereas the glycerol release level was not affected. When CG-1 was added into the medium during days 0-2 of 6-days-adipogenesis, the accumulation of intracellular lipids and the mRNA levels of the adipogenesis-related genes were decreased. In addition, the phosphorylation level of insulin receptor substrate-1 (IRS-1) and Akt in the early phase of adipocyte differentiation (within 1 day after initiating adipocyte differentiation) was reduced by CG-1. Furthermore, insulin-activated translocation of glucose transporter 4 to the plasma membranes in adipocytes was suppressed by CG-1, followed by decreased glucose uptake into the cells. These results indicate that an andiroba limonoid CG-1 suppressed the accumulation of intracellular lipids in the early phase of adipocyte differentiation through repression of IRS-1/Akt-mediated glucose uptake in adipocytes.
Collapse
Affiliation(s)
- Chihiro Matsumoto
- Department of Pathobiochemistry, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| | - Atsushi Koike
- Department of Pathobiochemistry, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| | - Reiko Tanaka
- Department of Medicinal Chemistry, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| | - Ko Fujimori
- Department of Pathobiochemistry, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| |
Collapse
|
50
|
Huber K, Hofer DC, Trefely S, Pelzmann HJ, Madreiter-Sokolowski C, Duta-Mare M, Schlager S, Trausinger G, Stryeck S, Graier WF, Kolb D, Magnes C, Snyder NW, Prokesch A, Kratky D, Madl T, Wellen KE, Bogner-Strauss JG. N-acetylaspartate pathway is nutrient responsive and coordinates lipid and energy metabolism in brown adipocytes. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2019; 1866:337-348. [PMID: 30595160 PMCID: PMC6390944 DOI: 10.1016/j.bbamcr.2018.08.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/27/2018] [Indexed: 12/22/2022]
Abstract
The discovery of significant amounts of metabolically active brown adipose tissue (BAT) in adult humans renders it a promising target for anti-obesity therapies by inducing weight loss through increased energy expenditure. The components of the N-acetylaspartate (NAA) pathway are highly abundant in BAT. Aspartate N-acetyltransferase (Asp-NAT, encoded by Nat8l) synthesizes NAA from acetyl-CoA and aspartate and increases energy expenditure in brown adipocytes. However, the exact mechanism how the NAA pathway contributes to accelerated mobilization and oxidation of lipids and the physiological regulation of the NAA pathway remained elusive. Here, we demonstrate that the expression of NAA pathway genes corresponds to nutrient availability and specifically responds to changes in exogenous glucose. NAA is preferentially produced from glucose-derived acetyl-CoA and aspartate and its concentration increases during adipogenesis. Overexpression of Nat8l drains glucose-derived acetyl-CoA into the NAA pool at the expense of cellular lipids and certain amino acids. Mechanistically, we elucidated that a combined activation of neutral and lysosomal (acid) lipolysis is responsible for the increased lipid degradation. Specifically, translocation of the transcription factor EB to the nucleus activates the biosynthesis of autophagosomes and lysosomes. Lipid degradation within lysosomes accompanied by adipose triglyceride lipase-mediated lipolysis delivers fatty acids for the support of elevated mitochondrial respiration. Together, our data suggest a crucial role of the NAA pathway in energy metabolism and metabolic adaptation in BAT.
Collapse
Affiliation(s)
- Katharina Huber
- Institute of Biochemistry, Graz University of Technology, Graz, Austria; Department of Cancer Biology, University of Pennsylvania, Philadelphia, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, USA
| | - Dina C Hofer
- Institute of Biochemistry, Graz University of Technology, Graz, Austria
| | - Sophie Trefely
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, USA; AJ Drexel Autism Institute, Drexel University, Philadelphia, USA
| | - Helmut J Pelzmann
- Institute of Biochemistry, Graz University of Technology, Graz, Austria
| | - Corina Madreiter-Sokolowski
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Cell Biology, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Madalina Duta-Mare
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Cell Biology, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Stefanie Schlager
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Cell Biology, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Gert Trausinger
- HEALTH Institute for Biomedicine and Health Sciences, Joanneum Research, Graz, Austria
| | - Sarah Stryeck
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Cell Biology, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Wolfgang F Graier
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Cell Biology, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Dagmar Kolb
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Christoph Magnes
- HEALTH Institute for Biomedicine and Health Sciences, Joanneum Research, Graz, Austria
| | | | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Cell Biology, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Cell Biology, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, USA
| | - Juliane G Bogner-Strauss
- Institute of Biochemistry, Graz University of Technology, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|