1
|
Hussain Y, R P, Empel C, Sharma D, Kloene L, Zhu WF, Kaiser A, Weizel L, Proschak E, Koenigs RM, Chauhan P. A Photochemical Strategy towards Michael Addition Reactions of Cyclopropenes. Angew Chem Int Ed Engl 2025; 64:e202416956. [PMID: 39509541 DOI: 10.1002/anie.202416956] [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: 09/03/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/15/2024]
Abstract
The development of Michael addition reactions to conjugated cyclopropenes is a challenge in organic synthesis due to the fleeting and reactive nature of such strained Michael acceptor systems. Herein, the development of a photochemical approach towards such conjugated cyclopropenes is reported that serves as a strategic entry point to densely functionalized cyclopropanes in a diastereoselective fashion. The process involves the light-mediated generation of transient cyclopropenyl α,β-unsaturated esters from vinyl diazo esters, followed by an organic base catalyzed nucleophilic addition of N-heterocycles to directly access β-N-heterocyclic cyclopropanoic esters. With this synergistic approach, various trisubstituted cyclopropanes bearing N-heteroaryl and N-heterocyclic rings such as indole, pyrrole, benzimidazole, isatin, pyridinone, and quinolinone were accessed efficiently in good yield and decent to good diastereoselectivities. Further, β-indolyl cyclopropanoic acids have been synthesized and were successfully evaluated as FABP-4 inhibitors. Theoretical calculations have been performed to elucidate the mechanism, which was further supported by experimental findings.
Collapse
Affiliation(s)
- Yaseen Hussain
- Department of Chemistry, Indian Institute of Technology Jammu, Jagti, NH-44, Nagrota Bypass, Jammu J&K, 181221, India
| | - Prasanna R
- Department of Chemistry, Indian Institute of Technology Jammu, Jagti, NH-44, Nagrota Bypass, Jammu J&K, 181221, India
| | - Claire Empel
- RWTH Aachen University, Institute of Organic Chemistry, Landoltweg 1, 52074, Aachen
| | - Deepak Sharma
- Department of Chemistry, Indian Institute of Technology Jammu, Jagti, NH-44, Nagrota Bypass, Jammu J&K, 181221, India
| | - Lennard Kloene
- RWTH Aachen University, Institute of Organic Chemistry, Landoltweg 1, 52074, Aachen
| | - W Felix Zhu
- Goethe University, Institute of Pharmaceutical Chemistry, Max-von-Laue Str. 9, 60438, Frankfurt am Main
| | - Astrid Kaiser
- Goethe University, Institute of Pharmaceutical Chemistry, Max-von-Laue Str. 9, 60438, Frankfurt am Main
| | - Lilia Weizel
- Goethe University, Institute of Pharmaceutical Chemistry, Max-von-Laue Str. 9, 60438, Frankfurt am Main
| | - Ewgenij Proschak
- Goethe University, Institute of Pharmaceutical Chemistry, Max-von-Laue Str. 9, 60438, Frankfurt am Main
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| | - Rene M Koenigs
- RWTH Aachen University, Institute of Organic Chemistry, Landoltweg 1, 52074, Aachen
| | - Pankaj Chauhan
- Department of Chemistry, Indian Institute of Technology Jammu, Jagti, NH-44, Nagrota Bypass, Jammu J&K, 181221, India
| |
Collapse
|
2
|
Numao S, Uchida R, Nakagaichi M. Response of circulating fatty acid binding protein 4 concentration to low-intensity acute aerobic exercise is amplified in an exercise duration-dependent manner in healthy men. J Physiol Anthropol 2024; 43:31. [PMID: 39707536 DOI: 10.1186/s40101-024-00379-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Circulating fatty acid-binding protein 4 (FABP4) influences cardiovascular disease and glucose metabolism. Acute aerobic exercise increases circulating FABP4 concentrations, but the factors underlying this effect in humans are unclear. We investigated the effect of exercise duration on circulating FABP4 concentrations in healthy men. METHODS This randomized crossover study enrolled healthy young men randomly assigned to two trials, short-duration (SE) and long-duration (LE) aerobic exercises trials. Both involved acute aerobic exercise followed by 60 min of bed rest. The exercise intensity was the same (40% peak oxygen uptake); however, the duration was 40 and 70 min for the SE and LE trials, respectively. Venous blood samples were collected to measure hormones, metabolites, and FABP4 concentrations. RESULTS Twelve healthy young men completed both trials. Changes in hormone levels did not differ significantly between the SE and LE trials (p > 0.05). However, the circulating FABP4 concentration increased significantly only in the LE trial immediately after exercise (p = 0.018). It increased significantly 30-60 min post-exercise in both the SE and LE trials (p < 0.018), with the extent of the increase being significantly higher in the LE trial than in the SE trial (p < 0.001). In each trial, the total incremental area under the curve of circulating FABP4 concentration was significantly positively correlated with body fat percentage (SE trial: rs = 0.699, p = 0.019; LE trial: rs = 0.643, p = 0.024). CONCLUSION Our findings suggest that exercise duration is associated with the magnitude of increased FABP4 secretion into the blood circulation. Body fat accumulation may also be involved in the magnitude of FABP4 secretion induced by acute aerobic exercise. TRIAL REGISTRATION The study was pre-registered with the University Hospital Medical Information Network Center (UMIN), a clinical trial registration system (ID: UMIN000051068).
Collapse
Affiliation(s)
- Shigeharu Numao
- Department of Sports and Life Sciences, National Institute of Fitness and Sports in Kanoya, 1 Shiromizu, Kanoya, Kagoshima, 891-2393, Japan.
| | - Ryota Uchida
- Health and Sports Promotion Division, Sports Innovation Organization, National Institute of Fitness and Sports in Kanoya, 1 Shiromizu, Kanoya, Kagoshima, 891-2393, Japan
| | - Masaki Nakagaichi
- Department of Sports and Life Sciences, National Institute of Fitness and Sports in Kanoya, 1 Shiromizu, Kanoya, Kagoshima, 891-2393, Japan
| |
Collapse
|
3
|
Masood A, Benabdelkamel H, Joy SS, Alhossan A, Alsuwayni B, Abdeen G, Aldhwayan M, Alfadda NA, Miras AD, Alfadda AA. Label-free quantitative proteomic profiling reveals differential plasma protein expression in patients with obesity after treatment with liraglutide. Front Mol Biosci 2024; 11:1458675. [PMID: 39324112 PMCID: PMC11422103 DOI: 10.3389/fmolb.2024.1458675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/12/2024] [Indexed: 09/27/2024] Open
Abstract
Introduction Treatment and management of obesity is clinically challenging. The inclusion of GLP-1 receptor agonists (GLP1RA) in the medical management of obesity has proven to be efficacious. However, mechanisms underlying the molecular changes arising from GLP1RA treatment in patients with obesity remain to be elucidated. Methods A single-center, prospective study was undertaken to evaluate the changes in the plasma proteins after liraglutide 3 mg therapy in twenty patients (M/F: 7/13) with obesity (mean BMI 40.65 ± 3.7 kg/m2). Anthropometric and laboratory parameters were measured, and blood samples were collected at two time points: baseline, before initiating treatment (pretreatment group, PT), and after three months of receiving the full dose liraglutide 3 mg (posttreatment group, PoT). An untargeted label-free LC MSMS mass spectrometric approach combined with bioinformatics and network pathway analysis was used to determine changes in the proteomic profiles. Results The mean age of the study participants was 36.0 ± 11.1 years. A statistically significant change was observed in weight, BMI and HbA1c levels between the PT and PoT groups (paired t-test, P < 0.001). A significant dysregulation was noted in the abundances of 151 proteins (31 up and 120 downregulated) between the two groups. The potential biomarkers were evaluated using receiver operating characteristic (ROC) curves. The top ten proteins (area under the curve (AUC) of 0.999 (95% CI)) were identified as potential biomarkers between PT and PoT groups and included Cystatin-B, major vault protein, and plastin-3, which were upregulated, whereas multimerin-2, large ribosomal P2, and proline-rich acidic protein 1 were downregulated in the PoT group compared with the PT group. The top network pathway identified using ingenuity pathway analysis (IPA), centered around dysregulation of MAPK, AKT, and PKc signaling pathways and related to cell-to-cell signaling and interaction, cellular assembly and organization, cellular compromise and a score of 46 with 25 focus proteins. Discussion Through label-free quantitative proteomic analysis, our study revealed significant dysregulation of plasma proteins after liraglutide 3 mg treatment in patients with obesity. The alterations in the proteomic profile between the PT and PoT groups demonstrated a decrease in levels of proteins involved in inflammation and oxidative stress pathways. On the other hand proteins involved in the glycolytic and lipolytic metabolic pathways as well as those participating in cytoskeletal and endothelial reorganization were observed to be increased. Understanding actions of liraglutide at a molecular and proteomic levels provides a holistic look into how liraglutide impacts metabolism, induces weight loss and improves overall metabolic health.
Collapse
Affiliation(s)
- Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Salini Scaria Joy
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdulaziz Alhossan
- Corporate of Pharmacy Services, King Saud University Medical City, Riyadh, Saudi Arabia
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Bashayr Alsuwayni
- Corporate of Pharmacy Services, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Ghalia Abdeen
- Department of Community Health Sciences, Clinical Nutrition, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Madhawi Aldhwayan
- Department of Community Health Sciences, Clinical Nutrition, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Nora A. Alfadda
- Department of Community Health Sciences, Clinical Nutrition, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Alexander Dimitri Miras
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolic Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
- School of Medicine, Ulster University, Derry, United Kingdom
| | - Assim A. Alfadda
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
4
|
Thillainadesan S, Lambert A, Cooke KC, Stöckli J, Yau B, Masson SWC, Howell A, Potter M, Fuller OK, Jiang YL, Kebede MA, Morahan G, James DE, Madsen S, Hocking SL. The metabolic consequences of 'yo-yo' dieting are markedly influenced by genetic diversity. Int J Obes (Lond) 2024; 48:1170-1179. [PMID: 38961153 PMCID: PMC11281900 DOI: 10.1038/s41366-024-01542-2] [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: 09/07/2023] [Revised: 04/09/2024] [Accepted: 05/10/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Weight loss can improve the metabolic complications of obesity. However, it is unclear whether insulin resistance persists despite weight loss and whether any protective benefits are preserved following weight regain (weight cycling). The impact of genetic background on weight cycling is undocumented. We aimed to investigate the effects of weight loss and weight cycling on metabolic outcomes and sought to clarify the role of genetics in this relationship. METHOD Both C57BL/6 J and genetically heterogeneous Diversity Outbred Australia (DOz) mice were alternately fed high fat Western-style diet (WD) and a chow diet at 8-week intervals. Metabolic measures including body composition, glucose tolerance, pancreatic beta cell activity, liver lipid levels and adipose tissue insulin sensitivity were determined. RESULTS After diet switch from WD (8-week) to chow (8-week), C57BL/6 J mice displayed a rapid normalisation of body weight, adiposity, hyperinsulinemia, liver lipid levels and glucose uptake into adipose tissue comparable to chow-fed controls. In response to the same dietary intervention, genetically diverse DOz mice conversely maintained significantly higher fat mass and insulin levels compared to chow-fed controls and exhibited much more profound interindividual variability than C57BL/6 J mice. Weight cycled (WC) animals were re-exposed to WD (8-week) and compared to age-matched controls fed 8-week WD for the first time (LOb). In C57BL/6 J but not DOz mice, WC animals had significantly higher blood insulin levels than LOb controls. All WC animals exhibited significantly greater beta cell activity than LOb controls despite similar fat mass, glucose tolerance, liver lipid levels and insulin-stimulated glucose uptake in adipose tissue. CONCLUSION Following weight loss, metabolic outcomes return to baseline in C57BL/6 J mice with obesity. However, genetic diversity significantly impacts this response. A period of weight loss does not provide lasting benefits after weight regain, and weight cycling is detrimental and associated with hyperinsulinemia and elevated basal insulin secretion.
Collapse
Affiliation(s)
- Senthil Thillainadesan
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Aaron Lambert
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Kristen C Cooke
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Jacqueline Stöckli
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Belinda Yau
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Stewart W C Masson
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Anna Howell
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Meg Potter
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Oliver K Fuller
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Yi Lin Jiang
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Melkam A Kebede
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Grant Morahan
- Australian Centre for Advancing Diabetes Innovations, Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - David E James
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia.
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, 2006, Australia.
| | - Søren Madsen
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, 2006, Australia.
| | - Samantha L Hocking
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia.
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia.
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
5
|
Alwan IH, Aljubouri TRS, Al-Shuhaib MBS. A Novel Missense SNP in the Fatty Acid-Binding Protein 4 (FABP4) Gene is Associated with Growth Traits in Karakul and Awassi Sheep. Biochem Genet 2024; 62:1462-1484. [PMID: 37640973 DOI: 10.1007/s10528-023-10504-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
The fatty acid-binding protein 4 (FABP4) plays a crucial role in the transportation and metabolism of fatty acids. It binds to long-chain fatty acids and facilitates their transport within cells. FABP4 is involved in lipid metabolism, insulin sensitivity, inflammation, and energy homeostasis. This study was conducted to assess the association between the FABP4 gene and growth traits in Karakul and Awassi sheep. A PCR-single strand conformation polymorphism (SSCP) protocol was utilized to assess the polymorphism of FABP4 PCR products with growth traits measured at birth, three, six, nine, and twelve-month intervals. One non-synonymous SNP was identified in the second exon, in which threonine was converted to aspartate in the 61st position in FABP4 (p.61Thr > Asp). This novel SNP showed significant associations with all growth traits measured at all age intervals. The results showed that lambs with the TT genotype exhibited higher growth traits than those with the GT and GG genotypes, respectively. The conducted prediction showed a clearly deleterious effect of p.61Thr > Asp on FABP4 structure, which was accompanied by reduced fatty acid binding efficiency. Owing to the predicted damaging effects caused by p.61Thr > Asp on FABP, lower levels of lipid transport and its consequent increased weight gain and other growth trait indices are expected. Therefore, a putative mechanism through which lambs with these genotypes exhibit higher growth traits is proposed. The FABP4 gene is suggested as a promising marker to improve growth traits in Karakul and Awassi sheep. However, more research is required to validate this mechanism.
Collapse
Affiliation(s)
- Ibrahim H Alwan
- Department of Animal Production, College of Agriculture, Al-Qasim Green University, Al-Qasim, Babil, 51001, Iraq
| | - Thamer R S Aljubouri
- Department of Animal Production, College of Agriculture, Al-Qasim Green University, Al-Qasim, Babil, 51001, Iraq
| | - Mohammed Baqur S Al-Shuhaib
- Department of Animal Production, College of Agriculture, Al-Qasim Green University, Al-Qasim, Babil, 51001, Iraq.
| |
Collapse
|
6
|
Bline AP, Ellis LB, Pelch KE, Lam J, Sen S, Zlatnik M, Varshavsky J. The effect of per and polyfluoroalkyl substance (PFAS) exposure on gestational diabetes mellitus and its subclinical risk factors: A systematic review and meta-analysis protocol. ENVIRONMENT INTERNATIONAL 2024; 188:108711. [PMID: 38754246 DOI: 10.1016/j.envint.2024.108711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/30/2024] [Accepted: 04/27/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Multiple lines of evidence suggest that exposure to per- and polyfluoroalkyl substances (PFAS) may alter glucose homeostasis, particularly during pregnancy, and may affect risk for developing gestational diabetes mellitus (GDM). While previous systematic reviews have been conducted on this topic, they did not assess internal validity of the included studies and their search strategies were narrowly focused. OBJECTIVE The objective of this study is to assess the effect of higher PFAS exposure (defined by individual compounds or mixtures measured before or during pregnancy) on GDM and subclinical measures of impaired glucose homeostasis (measured during pregnancy) compared to lower PFAS exposure in pregnant. METHODS We developed our systematic review protocol in accordance with the Navigation Guide. Peer-reviewed journal and grey literature searches were piloted in to identify relevant studies and refine our search terms and strategy. We also piloted the study screening criteria and data extraction form in DistillerSR, and refined our protocol accordingly. The risk of bias assessment protocol was adapted from Navigation Guide guidance and will be piloted and performed in DistillerSR. Pending the identification of comparable studies, quantitative meta-analyses will be performed where possible. Study results that cannot be quantitatively synthesized will be included in a narrative synthesis. The quality and strength of the body of evidence will be evaluated using Navigation Guide methodology, which is informed by guidance from the Cochrane Collaboration and Grading of Recommendations Assessment, Development and Evaluation (GRADE). We also made refinements to the quality of evidence considerations based on guidance from the National Institute of Environmental Health Sciences (NIEHS) Office of Health Assessment and Translation (OHAT). FUNDING This work was supported by the Systematizing Data on Per- and Polyfluoroalkyl Substances and Health Northeastern University TIER 1 Award.
Collapse
Affiliation(s)
- Abigail P Bline
- Social Science Environmental Health Research Institute, Northeastern University, Boston, MA, United States; Silent Spring Institute, Newton, MA, United States.
| | - Lauren B Ellis
- Social Science Environmental Health Research Institute, Northeastern University, Boston, MA, United States; Department of Health Sciences, Northeastern University, Boston, MA, United States.
| | - Katherine E Pelch
- Natural Resources Defense Council, San Francisco, CA, United States.
| | - Juleen Lam
- Department of Public Health, California State University, East Bay, Hayward, CA, United States.
| | - Saunak Sen
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States.
| | - Marya Zlatnik
- Department of Obstetrics, Gynecology and Reproductive Sciences, Program on Reproductive Health and the Environment, University of California San Francisco, San Francisco, CA, United States.
| | - Julia Varshavsky
- Social Science Environmental Health Research Institute, Northeastern University, Boston, MA, United States; Department of Health Sciences, Northeastern University, Boston, MA, United States; Department of Civil and Environmental Engineering, Northeastern University, Boston, MA, United States.
| |
Collapse
|
7
|
Khan A, Zahid MA, Mohammad A, Agouni A. Structure-guided engineering and molecular simulations to design a potent monoclonal antibody to target aP2 antigen for adaptive immune response instigation against type 2 diabetes. Front Immunol 2024; 15:1357342. [PMID: 38524133 PMCID: PMC10960362 DOI: 10.3389/fimmu.2024.1357342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 02/08/2024] [Indexed: 03/26/2024] Open
Abstract
Introduction Diabetes mellitus (DM) is recognized as one of the oldest chronic diseases and has become a significant public health issue, necessitating innovative therapeutic strategies to enhance patient outcomes. Traditional treatments have provided limited success, highlighting the need for novel approaches in managing this complex disease. Methods In our study, we employed graph signature-based methodologies in conjunction with molecular simulation and free energy calculations. The objective was to engineer the CA33 monoclonal antibody for effective targeting of the aP2 antigen, aiming to elicit a potent immune response. This approach involved screening a mutational landscape comprising 57 mutants to identify modifications that yield significant enhancements in binding efficacy and stability. Results Analysis of the mutational landscape revealed that only five substitutions resulted in noteworthy improvements. Among these, mutations T94M, A96E, A96Q, and T94W were identified through molecular docking experiments to exhibit higher docking scores compared to the wild-type. Further validation was provided by calculating the dissociation constant (KD), which showed a similar trend in favor of these mutations. Molecular simulation analyses highlighted T94M as the most stable complex, with reduced internal fluctuations upon binding. Principal components analysis (PCA) indicated that both the wild-type and T94M mutant displayed similar patterns of constrained and restricted motion across principal components. The free energy landscape analysis underscored a single metastable state for all complexes, indicating limited structural variability and potential for high therapeutic efficacy against aP2. Total binding free energy (TBE) calculations further supported the superior performance of the T94M mutation, with TBE values demonstrating the enhanced binding affinity of selected mutants over the wild-type. Discussion Our findings suggest that the T94M substitution, along with other identified mutations, significantly enhances the therapeutic potential of the CA33 antibody against DM by improving its binding affinity and stability. These results not only contribute to a deeper understanding of antibody-antigen interactions in the context of DM but also provide a valuable framework for the rational design of antibodies aimed at targeting this disease more effectively.
Collapse
Affiliation(s)
- Abbas Khan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Muhammad Ammar Zahid
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Anwar Mohammad
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
8
|
Numao S, Uchida R, Kurosaki T, Nakagaichi M. Carbohydrate ingestion does not suppress increases in fatty acid-binding protein 4 concentrations post-acute aerobic exercise in healthy men: a randomized crossover study. BMC Sports Sci Med Rehabil 2024; 16:63. [PMID: 38438929 PMCID: PMC10913278 DOI: 10.1186/s13102-024-00852-2] [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: 04/24/2023] [Accepted: 02/19/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND Fatty acid-binding protein 4 (FABP4) has been associated with cardiovascular disease and diabetes. Acute aerobic exercise increases circulating FABP4 concentrations, but the underlying mechanisms remain unclear. The purpose of this study was to investigate the effects of inhibition of lipolysis by carbohydrate ingestion on circulating FABP4 concentrations during and after acute aerobic exercise in healthy men. METHODS Men aged between 20 and 40, with no exercise habits and no metabolic diseases, were recruited. In a randomized crossover design, the participants underwent a carbohydrate-ingestion exercise (CE) and a fasted exercise (FE) trial. The CE trial consisted of 40-min acute aerobic exercise with ingestion of carbohydrates and 60-min bed rest. The FE trial followed the same protocol as the CE trial but without carbohydrate ingestion. Venous blood samples were collected to measure hormones (adrenaline, noradrenaline, and insulin) metabolites (glycerol, free fatty acids, and glucose), and FABP4 concentrations. Ventilation and gas exchange were also collected to measure substrate oxidation. RESULTS Thirteen healthy men participated in and completed both the CE and FE trials. The insulin concentration was more than 4 times higher in the CE trial than in the FE trial (p < 0.004, effect size [ES] > 2.00). Free fatty acid concentrations were more than 4 times lower in the CE trial than in the FE trial (p < 0.02, ES > 2.04). However, there was no significant difference in the changes in circulating FABP4 concentrations between the CE and FE trials (p = 0.108), which did not change during aerobic exercise and significantly increased post-aerobic exercise in both trials (p < 0.002, ES > 1.212). Changes in FABP4 concentrations following aerobic exercise were not significantly correlated with changes in glycerol or free fatty acid concentrations during aerobic exercise. CONCLUSIONS The results suggest that suppression of lipolysis and elevation of insulin are not strongly involved in increases in FABP4 secretion following acute aerobic exercise.
Collapse
Affiliation(s)
- Shigeharu Numao
- National Institute of Fitness and Sports in Kanoya, 1 Shiromizu, 891-2393, Kanoya, Kagoshima, Japan.
| | - Ryota Uchida
- National Institute of Fitness and Sports in Kanoya, 1 Shiromizu, 891-2393, Kanoya, Kagoshima, Japan
| | - Takashi Kurosaki
- National Institute of Fitness and Sports in Kanoya, 1 Shiromizu, 891-2393, Kanoya, Kagoshima, Japan
| | - Masaki Nakagaichi
- National Institute of Fitness and Sports in Kanoya, 1 Shiromizu, 891-2393, Kanoya, Kagoshima, Japan
| |
Collapse
|
9
|
Mallick R, Basak S, Das RK, Banerjee A, Paul S, Pathak S, Duttaroy AK. Fatty Acids and their Proteins in Adipose Tissue Inflammation. Cell Biochem Biophys 2024; 82:35-51. [PMID: 37794302 PMCID: PMC10867084 DOI: 10.1007/s12013-023-01185-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/06/2023]
Abstract
Chronic low-grade adipose tissue inflammation is associated with metabolic disorders. Inflammation results from the intertwined cross-talks of pro-inflammatory and anti-inflammatory pathways in the immune response of adipose tissue. In addition, adipose FABP4 levels and lipid droplet proteins are involved in systemic and tissue inflammation. Dysregulated adipocytes help infiltrate immune cells derived from bone marrow responsible for producing cytokines and chemokines. When adipose tissue expands in excess, adipocyte exhibits increased secretion of adipokines and is implicated in metabolic disturbances due to the release of free fatty acids. This review presents an emerging concept in adipose tissue fat metabolism, fatty acid handling and binding proteins, and lipid droplet proteins and their involvement in inflammatory disorders.
Collapse
Affiliation(s)
- Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Ranjit K Das
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Antara Banerjee
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc, San Pablo, Queretaro, 76130, Mexico
| | - Surajit Pathak
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, POB 1046 Blindern, Oslo, Norway.
| |
Collapse
|
10
|
Gilani A, Stoll L, Homan EA, Lo JC. Adipose Signals Regulating Distal Organ Health and Disease. Diabetes 2024; 73:169-177. [PMID: 38241508 PMCID: PMC10796297 DOI: 10.2337/dbi23-0005] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/03/2023] [Indexed: 01/21/2024]
Abstract
Excessive adiposity in obesity is a significant risk factor for development of type 2 diabetes (T2D), nonalcoholic fatty liver disease, and other cardiometabolic diseases. An unhealthy expansion of adipose tissue (AT) results in reduced adipogenesis, increased adipocyte hypertrophy, adipocyte hypoxia, chronic low-grade inflammation, increased macrophage infiltration, and insulin resistance. This ultimately culminates in AT dysfunction characterized by decreased secretion of antidiabetic adipokines such as adiponectin and adipsin and increased secretion of proinflammatory prodiabetic adipokines including RBP4 and resistin. This imbalance in adipokine secretion alters the physiological state of AT communication with target organs including pancreatic β-cells, heart, and liver. In the pancreatic β-cells, adipokines are known to have a direct effect on insulin secretion, gene expression, cell death, and/or dedifferentiation. For instance, impaired secretion of adipsin, which promotes insulin secretion and β-cell identity, results in β-cell failure and T2D, thus presenting a potential druggable target to improve and/or preserve β-cell function. The cardiac tissue is affected by both the classic white AT-secreted adipokines and the newly recognized brown AT (BAT)-secreted BATokines or lipokines that alter lipid deposition and ventricular function. In the liver, adipokines affect hepatic gluconeogenesis, lipid accumulation, and insulin sensitivity, underscoring the importance of adipose-liver communication in the pathogenesis of nonalcoholic fatty liver disease. In this perspective, we outline what is currently known about the effects of individual adipokines on pancreatic β-cells, liver, and the heart.
Collapse
Affiliation(s)
- Ankit Gilani
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Lisa Stoll
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Edwin A. Homan
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - James C. Lo
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY
| |
Collapse
|
11
|
Avtanski D, Stojchevski R. Significance of Adipose Tissue as an Endocrine Organ. CONTEMPORARY ENDOCRINOLOGY 2024:1-46. [DOI: 10.1007/978-3-031-72570-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Yu HC, Jeon YG, Na AY, Han CY, Lee MR, Yang JD, Yu HC, Son JB, Kim ND, Kim JB, Lee S, Bae EJ, Park BH. p21-activated kinase 4 counteracts PKA-dependent lipolysis by phosphorylating FABP4 and HSL. Nat Metab 2024; 6:94-112. [PMID: 38216738 DOI: 10.1038/s42255-023-00957-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 11/30/2023] [Indexed: 01/14/2024]
Abstract
Adipose tissue lipolysis is mediated by cAMP-protein kinase A (PKA)-dependent intracellular signalling. Here, we show that PKA targets p21-activated kinase 4 (PAK4), leading to its protein degradation. Adipose tissue-specific overexpression of PAK4 in mice attenuates lipolysis and exacerbates diet-induced obesity. Conversely, adipose tissue-specific knockout of Pak4 or the administration of a PAK4 inhibitor in mice ameliorates diet-induced obesity and insulin resistance while enhancing lipolysis. Pak4 knockout also increases energy expenditure and adipose tissue browning activity. Mechanistically, PAK4 directly phosphorylates fatty acid-binding protein 4 (FABP4) at T126 and hormone-sensitive lipase (HSL) at S565, impairing their interaction and thereby inhibiting lipolysis. Levels of PAK4 and the phosphorylation of FABP4-T126 and HSL-S565 are enhanced in the visceral fat of individuals with obesity compared to their lean counterparts. In summary, we have uncovered an important role for FABP4 phosphorylation in regulating adipose tissue lipolysis, and PAK4 inhibition may offer a therapeutic strategy for the treatment of obesity.
Collapse
Affiliation(s)
- Hwang Chan Yu
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju, Korea
| | - Yong Geun Jeon
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Ann-Yae Na
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Chang Yeob Han
- School of Pharmacy, Jeonbuk National University, Jeonju, Korea
| | - Mi Rin Lee
- Department of Surgery, Jeonbuk National University Hospital, Jeonju, Korea
| | - Jae Do Yang
- Department of Surgery, Jeonbuk National University Hospital, Jeonju, Korea
| | - Hee Chul Yu
- Department of Surgery, Jeonbuk National University Hospital, Jeonju, Korea
| | | | | | - Jae Bum Kim
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Sangkyu Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea.
| | - Eun Ju Bae
- School of Pharmacy, Jeonbuk National University, Jeonju, Korea.
| | - Byung-Hyun Park
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju, Korea.
| |
Collapse
|
13
|
Abstract
Fatty acid-binding proteins (FABPs) are small lipid-binding proteins abundantly expressed in tissues that are highly active in fatty acid (FA) metabolism. Ten mammalian FABPs have been identified, with tissue-specific expression patterns and highly conserved tertiary structures. FABPs were initially studied as intracellular FA transport proteins. Further investigation has demonstrated their participation in lipid metabolism, both directly and via regulation of gene expression, and in signaling within their cells of expression. There is also evidence that they may be secreted and have functional impact via the circulation. It has also been shown that the FABP ligand binding repertoire extends beyond long-chain FAs and that their functional properties also involve participation in systemic metabolism. This article reviews the present understanding of FABP functions and their apparent roles in disease, particularly metabolic and inflammation-related disorders and cancers.
Collapse
Affiliation(s)
- Judith Storch
- Department of Nutritional Sciences and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, United States;
| | - Betina Corsico
- Instituto de Investigaciones Bioquímicas de La Plata, CONICET-UNLP, Facultad de Ciencias Médicas, La Plata, Argentina;
| |
Collapse
|
14
|
Chen D, Wirth KM, Kizy S, Muretta JM, Markowski TW, Yong P, Sheka A, Abdelwahab H, Hertzel AV, Ikramuddin S, Yamamoto M, Bernlohr DA. Desmoglein 2 Functions as a Receptor for Fatty Acid Binding Protein 4 in Breast Cancer Epithelial Cells. Mol Cancer Res 2023; 21:836-848. [PMID: 37115197 PMCID: PMC10524127 DOI: 10.1158/1541-7786.mcr-22-0763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/19/2023] [Accepted: 04/26/2023] [Indexed: 04/29/2023]
Abstract
Fatty acid binding protein 4 (FABP4) is a secreted adipokine linked to obesity and progression of a variety of cancers. Obesity increases extracellular FABP4 (eFABP4) levels in animal models and in obese breast cancer patients compared with lean healthy controls. Using MCF-7 and T47D breast cancer epithelial cells, we show herein that eFABP4 stimulates cellular proliferation in a time and concentration dependent manner while the non-fatty acid-binding mutant, R126Q, failed to potentiate growth. When E0771 murine breast cancer cells were injected into mice, FABP4 null animals exhibited delayed tumor growth and enhanced survival compared with injections into control C57Bl/6J animals. eFABP4 treatment of MCF-7 cells resulted in a significant increase in phosphorylation of extracellular signal-regulated kinase 1/2 (pERK), transcriptional activation of nuclear factor E2-related factor 2 (NRF2) and corresponding gene targets ALDH1A1, CYP1A1, HMOX1, SOD1 and decreased oxidative stress, while R126Q treatment did not show any effects. Proximity-labeling employing an APEX2-FABP4 fusion protein revealed several proteins functioning in desmosomes as eFABP4 receptor candidates including desmoglein (DSG), desmocollin, junction plankoglobin, desomoplankin, and cytokeratins. AlphaFold modeling predicted an interaction between eFABP4, and the extracellular cadherin repeats of DSG2 and pull-down and immunoprecipitation assays confirmed complex formation that was potentiated by oleic acid. Silencing of DSG2 in MCF-7 cells attenuated eFABP4 effects on cellular proliferation, pERK levels, and ALDH1A1 expression compared with controls. IMPLICATIONS These results suggest desmosomal proteins, and in particular desmoglein 2, may function as receptors of eFABP4 and provide new insight into the development and progression of obesity-associated cancers.
Collapse
Affiliation(s)
- Dongmei Chen
- Department of Biochemistry, Molecular Biology and Biophysics, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - Keith M. Wirth
- Department of Surgery, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - Scott Kizy
- Department of Surgery, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - Joseph M. Muretta
- Department of Biochemistry, Molecular Biology and Biophysics, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - Todd W Markowski
- Department of Biochemistry, Molecular Biology and Biophysics, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - Peter Yong
- Department of Biochemistry, Molecular Biology and Biophysics, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - Adam Sheka
- Department of Surgery, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - Hisham Abdelwahab
- Department of Surgery, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - Ann V. Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - Sayeed Ikramuddin
- Department of Surgery, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - Masato Yamamoto
- Department of Surgery, The University of Minnesota-Twin Cities, Minneapolis, MN USA
- Department of Masonic Cancer Center, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, The University of Minnesota-Twin Cities, Minneapolis, MN USA
| |
Collapse
|
15
|
Kumar S, Chhimwal J, Kumar S, Singh R, Patial V, Purohit R, Padwad YS. Phloretin and phlorizin mitigates inflammatory stress and alleviate adipose and hepatic insulin resistance by abrogating PPARγ S273-Cdk5 interaction in type 2 diabetic mice. Life Sci 2023; 322:121668. [PMID: 37023949 DOI: 10.1016/j.lfs.2023.121668] [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: 03/01/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
AIMS The rising prevalence of type 2 diabetes mellitus (T2DM) and accompanying insulin resistance is alarming globally. Natural and synthetic agonists of PPARγ are potentially attractive candidates for diabetics and are known to efficiently reverse adipose and hepatic insulin resistance, but related side effects and escalating costs are the causes of concern. Therefore, targeting PPARγ with natural ligands is advantageous and promising approach for the better management of T2DM. The present research aimed to assess the antidiabetic potential of phenolics Phloretin (PTN) and Phlorizin (PZN) in type 2 diabetic mice. MAIN METHODS In silico docking was performed to check the effect of PTN and PZN on PPARγ S273-Cdk5 interactions. The docking results were further validated in preclinical settings by utilizing a mice model of high fat diet-induced T2DM. KEY FINDINGS Computational docking and further MD-simulation data revealed that PTN and PZN inhibited the activation of Cdk5, thereby blocking the phosphorylation of PPARγ. Our in vivo results further demonstrated that PTN and PZN administration significantly improved the secretory functions of adipocytes by increasing adiponectin and reducing inflammatory cytokine levels, which ultimately reduced the hyperglycaemic index. Additionally, combined treatment of PTN and PZN decreased in vivo adipocyte expansion and increased Glut4 expression in adipose tissues. Furthermore, PTN and PZN treatment reduced hepatic insulin resistance by modulating lipid metabolism and inflammatory markers. SIGNIFICANCE In summary, our findings strongly imply that PTN and PZN are candidates as nutraceuticals in the management of comorbidities related to diabetes and its complications.
Collapse
Affiliation(s)
- Shiv Kumar
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Jyoti Chhimwal
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Suresh Kumar
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Rahul Singh
- Structural Bioinformatics Lab, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Rituraj Purohit
- Structural Bioinformatics Lab, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India.
| | - Yogendra S Padwad
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
16
|
Burkhardt LM, Bucher CH, Löffler J, Rinne C, Duda GN, Geissler S, Schulz TJ, Schmidt-Bleek K. The benefits of adipocyte metabolism in bone health and regeneration. Front Cell Dev Biol 2023; 11:1104709. [PMID: 36895792 PMCID: PMC9988968 DOI: 10.3389/fcell.2023.1104709] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Patients suffering from musculoskeletal diseases must cope with a diminished quality of life and an increased burden on medical expenses. The interaction of immune cells and mesenchymal stromal cells during bone regeneration is one of the key requirements for the restoration of skeletal integrity. While stromal cells of the osteo-chondral lineage support bone regeneration, an excessive accumulation of cells of the adipogenic lineage is thought to promote low-grade inflammation and impair bone regeneration. Increasing evidence indicates that pro-inflammatory signaling from adipocytes is responsible for various chronic musculoskeletal diseases. This review aims to summarize the features of bone marrow adipocytes by phenotype, function, secretory features, metabolic properties and their impact on bone formation. In detail, the master regulator of adipogenesis and prominent diabetes drug target, peroxisome proliferator-activated receptor γ (PPARG), will be debated as a potential therapeutic approach to enhance bone regeneration. We will explore the possibilities of using clinically established PPARG agonists, the thiazolidinediones (TZDs), as a treatment strategy to guide the induction of a pro-regenerative, metabolically active bone marrow adipose tissue. The impact of this PPARG induced bone marrow adipose tissue type on providing the necessary metabolites to sustain osteogenic-as well as beneficial immune cells during bone fracture healing will be highlighted.
Collapse
Affiliation(s)
- Lisa-Marie Burkhardt
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Christian H Bucher
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Julia Löffler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Charlotte Rinne
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Sven Geissler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
17
|
Sahu B, Bal NC. Adipokines from white adipose tissue in regulation of whole body energy homeostasis. Biochimie 2023; 204:92-107. [PMID: 36084909 DOI: 10.1016/j.biochi.2022.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/08/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023]
Abstract
Diseases originating from altered energy homeostasis including obesity, and type 2 diabetes are rapidly increasing worldwide. Research in the last few decades on animal models and humans demonstrates that the white adipose tissue (WAT) is critical for energy balance and more than just an energy storage site. WAT orchestrates the whole-body metabolism through inter-organ crosstalk primarily mediated by cytokines named "Adipokines". The adipokines influence metabolism and fuel selection of the skeletal muscle and liver thereby fine-tuning the load on WAT itself in physiological conditions like starvation, exercise and cold. In addition, adipokine secretion is influenced by various pathological conditions like obesity, inflammation and diabetes. In this review, we have surveyed the current state of knowledge on important adipokines and their significance in regulating energy balance and metabolic diseases. Furthermore, we have summarized the interplay of pro-inflammatory and anti-inflammatory adipokines in the modulation of pathological conditions.
Collapse
Affiliation(s)
- Bijayashree Sahu
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India.
| | - Naresh C Bal
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
18
|
Liu S, Wu D, Fan Z, Yang J, Li Y, Meng Y, Gao C, Zhan H. FABP4 in obesity-associated carcinogenesis: Novel insights into mechanisms and therapeutic implications. Front Mol Biosci 2022; 9:973955. [PMID: 36060264 PMCID: PMC9438896 DOI: 10.3389/fmolb.2022.973955] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 11/23/2022] Open
Abstract
The increasing prevalence of obesity worldwide is associated with an increased risk of various diseases, including multiple metabolic diseases, cardiovascular diseases, and malignant tumors. Fatty acid binding proteins (FABPs) are members of the adipokine family of multifunctional proteins that are related to fatty acid metabolism and are divided into 12 types according to their tissue origin. FABP4 is mainly secreted by adipocytes and macrophages. Under obesity, the synthesis of FABP4 increases, and the FABP4 content is higher not only in tissues but also in the blood, which promotes the occurrence and development of various cancers. Here, we comprehensively investigated obesity epidemiology and the biological mechanisms associated with the functions of FABP4 that may explain this effect. In this review, we explore the molecular mechanisms by which FABP4 promotes carcinoma development and the interaction between fat and cancer cells in obese circumstances here. This review leads us to understand how FABP4 signaling is involved in obesity-associated tumors, which could increase the potential for advancing novel therapeutic strategies and molecular targets for the systematic treatment of malignant tumors.
Collapse
|
19
|
Reiterer M, Gilani A, Lo JC. Pancreatic Islets as a Target of Adipokines. Compr Physiol 2022; 12:4039-4065. [PMID: 35950650 DOI: 10.1002/cphy.c210044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Rising rates of obesity are intricately tied to the type 2 diabetes epidemic. The adipose tissues can play a central role in protection against or triggering metabolic diseases through the secretion of adipokines. Many adipokines may improve peripheral insulin sensitivity through a variety of mechanisms, thereby indirectly reducing the strain on beta cells and thus improving their viability and functionality. Such effects will not be the focus of this article. Rather, we will focus on adipocyte-secreted molecules that have a direct effect on pancreatic islets. By their nature, adipokines represent potential druggable targets that can reach the islets and improve beta-cell function or preserve beta cells in the face of metabolic stress. © 2022 American Physiological Society. Compr Physiol 12:1-27, 2022.
Collapse
Affiliation(s)
- Moritz Reiterer
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - James C Lo
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
20
|
Huang J, Chen G, Zhang Q, Wang Y, Meng Q, Xu F, Zhang X, Zou W, Mi F, Yin J. Correlation between adipocyte fatty acid binding protein and glucose dysregulation is closely associated with obesity and metabolic syndrome: A cohort of Han Chinese population from Yunnan plateau. Lipids 2022; 57:257-264. [PMID: 35778866 DOI: 10.1002/lipd.12353] [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: 04/14/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 11/09/2022]
Abstract
The present study investigated the correlation of plasma A-FABP with glucose dysregulation under different body mass index (BMI) and metabolic states in a Han Chinese population from Yunnan plateau. We cross-sectionally analyzed data from the China Multi Ethnic Cohort, Yunnan province. Participants were divided into two groups. Group A contained 297 obese individuals with metabolic syndrome (MetS). Group B contained 326 age-, sex-, and region-matched normal BMI subjects without MetS. Glucose dysregulation was defined as elevated fasting plasma glucose (FPG) (FPG ≥ 5.6 mmol/L or current use of oral hypoglycemic agents or insulin). Circulating A-FABP were assayed by ELISA method. Binary and multiple regression analyses were preformed to evaluate the correlation between A-FABP and glucose dysregulation. Plasma A-FABP level was significantly higher in group A compared with group B (p < 0.001). Plasma A-FABP level correlated positively with elevated FPG in group A (r = 0.120, p = 0.039), but negatively with elevated FPG in group B (r = -0.115, p = 0.039). Multiple logistic regression analysis revealed that A-FABP was an independent predictor for elevated FPG in group A (β, 0.028; 95% CI, 1.001-1.056; p < 0.05), but not in group B (β, -0.008; 95% CI, 0.882-1.117; p > 0.05). In this study, A-FABP was an independent risk factor for glucose dysregulation in obese individuals with MetS living in the Yunnan plateau, but not for those without obesity and MetS.
Collapse
Affiliation(s)
- Juan Huang
- School of Public Health, Kunming Medical University, Kunming, China
| | - Guo Chen
- School of Public Health, Kunming Medical University, Kunming, China
| | - Qiao Zhang
- School of Public Health, Kunming Medical University, Kunming, China
| | - Yanjiao Wang
- School of Public Health, Kunming Medical University, Kunming, China
| | - Qiong Meng
- School of Public Health, Kunming Medical University, Kunming, China
| | - Fang Xu
- School of Public Health, Kunming Medical University, Kunming, China
| | - Xuehui Zhang
- School of Public Health, Kunming Medical University, Kunming, China
| | - Wei Zou
- School of Public Health, Kunming Medical University, Kunming, China
| | - Fei Mi
- School of Public Health, Kunming Medical University, Kunming, China
| | - Jianzhong Yin
- School of Public Health, Kunming Medical University, Kunming, China.,Baoshan College of Traditional Chinese Medicine, Baoshan, China
| |
Collapse
|
21
|
Vorobjova T, Tagoma A, Talja I, Janson H, Kirss A, Uibo R. FABP4 and I-FABP Levels in Pregnant Women Are Associated with Body Mass Index but Not Gestational Diabetes. J Diabetes Res 2022; 2022:1089434. [PMID: 35647197 PMCID: PMC9142318 DOI: 10.1155/2022/1089434] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Gestational diabetes mellitus (GDM) is glucose intolerance detected initially during pregnancy. GDM poses an increased risk for the development of diabetes later in life. Fatty acid-binding protein 4 (FABP4) is a regulator of lipid metabolism and is associated with obesity, insulin resistance, and type 2 diabetes. Increased level of intestinal fatty acid-binding protein (I-FABP) may indicate impaired intestinal permeability, which may be an important contributor to the pathogenesis of type 1 diabetes and GDM. We aimed to compare FABP4 and I-FABP levels in pregnant women with GDM and in healthy pregnant controls, taking into consideration their prepregnancy body mass index (BMI), past exposures to enteroviruses (EV), and adipokine and cytokine levels, which have been shown to decrease insulin sensitivity. Material and Methods. Forty patients with GDM (median age 30.5) and 40 pregnant healthy controls (median age 31.1) were divided on the basis of their prepregnancy BMI into two groups: normal weight (BMI < 25, n = 20) and overweight (BMI ≥ 25, n = 20). FABP4 and I-FABP were measured from serum samples using commercial ELISA kits. RESULTS FABP4 and I-FABP levels did not differ between women with GDM and healthy pregnant controls (p > 0.05 for both comparisons). However, both levels were associated with BMI (p < 0.001 for both comparisons). Median I-FABP level was the highest in healthy controls with lower BMI (<25) (p = 0.0009). FABP4 levels correlated with BMI and C-peptide values in both groups (p < 0.001). Anti-EV antibody levels did not correlate with FABP4 or I-FABP levels. FABP4 and adiponectin levels were negatively correlated in controls (r = -0.61, p = 0.0009), while I-FABP correlated positively with adiponectin (r = 0.58, p = 0.04) and resistin (r = 0.67, p = 0.04) levels in the GDM group. CONCLUSION FABP4 and I-FABP levels were not dependent on the diagnosis of GDM, but rather on BMI. The correlation of I-FABP with adiponectin and resistin levels in women with GDM may suggests the importance of lipid metabolism in GDM-associated changes in intestinal permeability.
Collapse
Affiliation(s)
- Tamara Vorobjova
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Aili Tagoma
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Ija Talja
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Helis Janson
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Anne Kirss
- Women's Clinic, Tartu University Hospital, L. Puusepa 8, Tartu 51014, Estonia
| | - Raivo Uibo
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| |
Collapse
|
22
|
Biondi G, Marrano N, Borrelli A, Rella M, Palma G, Calderoni I, Siciliano E, Lops P, Giorgino F, Natalicchio A. Adipose Tissue Secretion Pattern Influences β-Cell Wellness in the Transition from Obesity to Type 2 Diabetes. Int J Mol Sci 2022; 23:ijms23105522. [PMID: 35628332 PMCID: PMC9143684 DOI: 10.3390/ijms23105522] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 12/10/2022] Open
Abstract
The dysregulation of the β-cell functional mass, which is a reduction in the number of β-cells and their ability to secure adequate insulin secretion, represents a key mechanistic factor leading to the onset of type 2 diabetes (T2D). Obesity is recognised as a leading cause of β-cell loss and dysfunction and a risk factor for T2D. The natural history of β-cell failure in obesity-induced T2D can be divided into three steps: (1) β-cell compensatory hyperplasia and insulin hypersecretion, (2) insulin secretory dysfunction, and (3) loss of β-cell mass. Adipose tissue (AT) secretes many hormones/cytokines (adipokines) and fatty acids that can directly influence β-cell function and viability. As this secretory pattern is altered in obese and diabetic patients, it is expected that the cross-talk between AT and pancreatic β-cells could drive the maintenance of the β-cell integrity under physiological conditions and contribute to the reduction in the β-cell functional mass in a dysmetabolic state. In the current review, we summarise the evidence of the ability of the AT secretome to influence each step of β-cell failure, and attempt to draw a timeline of the alterations in the adipokine secretion pattern in the transition from obesity to T2D that reflects the progressive deterioration of the β-cell functional mass.
Collapse
|
23
|
Basak S, Banerjee A, Pathak S, Duttaroy AK. Dietary Fats and the Gut Microbiota: Their impacts on lipid-induced metabolic syndrome. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
24
|
Jiang Y, Han X, Feng N, Jin W, Zhang T, Zhang M, Shi M, Zhang B, Liu S, Hu D. Androgen plays an important role in regulating the synthesis of pheromone in the scent gland of muskrat. J Steroid Biochem Mol Biol 2022; 217:106026. [PMID: 34808361 DOI: 10.1016/j.jsbmb.2021.106026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/14/2021] [Accepted: 11/18/2021] [Indexed: 02/04/2023]
Abstract
The scent (musk) gland is an organ unique to muskrats and other scent-secreting animals, and the pheromones (musk) synthesized and secreted by the scent gland play a role in chemical communication among scent-secreting animals. The musk gland is synchronized with testicular developmental changes; however, little is known regarding androgen secretion from the testis and how this regulates pheromone synthesis and the secretion of scent. To investigate the effect of androgens on the synthesis of pheromones in the musk gland, we established a muskrat castration model by surgical removal of the testis, and analyzed the histomorphology, hormone concentration, gene expression, and changes in the chemical composition of the musk gland in castration and control groups by histomorphological analysis, Enzyme-Linked ImmunoSorbent Assay (ELISA), RNA sequencing (RNA-seq), and gas chromatography-mass spectrometry (GCMS). Histomorphological analysis results showed that after castration, muskrat gland cells underwent significant atrophy (P < 0.05). Hormone measurement results showed that there was a significant decrease in serum testosterone and muskrat musk testosterone (P < 0.05) after muskrat castration. Transcriptome sequencing results showed that 510 differentially expressed transcripts (DETs) were mainly enriched in fatty acid metabolism, terpenoid backbone biosynthesis, fatty acid degradation, PPAR signaling pathway, and fatty acid biosynthesis. GCMS results showed that macrocyclic ketones, steroids, fatty acids, alcohols, and esters in musk were significantly changed (P < 0.05). In conclusion, androgens were found to play an important function in the chemical communication exchange between muskrats through regulating pheromone synthesis in musk cells. This study provides a basis for understanding the mechanism of animal communication influenced by androgens.
Collapse
Affiliation(s)
- Yuanlin Jiang
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Xiangyu Han
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Nuannuan Feng
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Weijiang Jin
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Tianxiang Zhang
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Meishan Zhang
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Minghui Shi
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Baofeng Zhang
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Shuqiang Liu
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, 100083, People's Republic of China.
| | - Defu Hu
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, 100083, People's Republic of China.
| |
Collapse
|
25
|
NUMAO S, URITA Y, MATSUMURA I, TAKAI Y, UCHIDA R, KUROSAKI T, NAKAGAICHI M. Difference in circulating fatty acid binding protein 4 concentration in trained men. GAZZETTA MEDICA ITALIANA ARCHIVIO PER LE SCIENZE MEDICHE 2022. [DOI: 10.23736/s0393-3660.21.04589-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
26
|
Ren Y, Zhao H, Yin C, Lan X, Wu L, Du X, Griffiths HR, Gao D. Adipokines, Hepatokines and Myokines: Focus on Their Role and Molecular Mechanisms in Adipose Tissue Inflammation. Front Endocrinol (Lausanne) 2022; 13:873699. [PMID: 35909571 PMCID: PMC9329830 DOI: 10.3389/fendo.2022.873699] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/17/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic low-grade inflammation in adipose tissue (AT) is a hallmark of obesity and contributes to various metabolic disorders, such as type 2 diabetes and cardiovascular diseases. Inflammation in ATs is characterized by macrophage infiltration and the activation of inflammatory pathways mediated by NF-κB, JNK, and NLRP3 inflammasomes. Adipokines, hepatokines and myokines - proteins secreted from AT, the liver and skeletal muscle play regulatory roles in AT inflammation via endocrine, paracrine, and autocrine pathways. For example, obesity is associated with elevated levels of pro-inflammatory adipokines (e.g., leptin, resistin, chemerin, progranulin, RBP4, WISP1, FABP4, PAI-1, Follistatin-like1, MCP-1, SPARC, SPARCL1, and SAA) and reduced levels of anti-inflammatory adipokines such as adiponectin, omentin, ZAG, SFRP5, CTRP3, vaspin, and IL-10. Moreover, some hepatokines (Fetuin A, DPP4, FGF21, GDF15, and MANF) and myokines (irisin, IL-6, and DEL-1) also play pro- or anti-inflammatory roles in AT inflammation. This review aims to provide an updated understanding of these organokines and their role in AT inflammation and related metabolic abnormalities. It serves to highlight the molecular mechanisms underlying the effects of these organokines and their clinical significance. Insights into the roles and mechanisms of these organokines could provide novel and potential therapeutic targets for obesity-induced inflammation.
Collapse
Affiliation(s)
- Yakun Ren
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
| | - Hao Zhao
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xi Lan
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Litao Wu
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Xiaojuan Du
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Helen R. Griffiths
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Dan Gao
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
- *Correspondence: Dan Gao,
| |
Collapse
|
27
|
Prentice KJ, Saksi J, Robertson LT, Lee GY, Inouye KE, Eguchi K, Lee A, Cakici O, Otterbeck E, Cedillo P, Achenbach P, Ziegler AG, Calay ES, Engin F, Hotamisligil GS. A hormone complex of FABP4 and nucleoside kinases regulates islet function. Nature 2021; 600:720-726. [PMID: 34880500 DOI: 10.1038/s41586-021-04137-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/14/2021] [Indexed: 11/09/2022]
Abstract
The liberation of energy stores from adipocytes is critical to support survival in times of energy deficit; however, uncontrolled or chronic lipolysis associated with insulin resistance and/or insulin insufficiency disrupts metabolic homeostasis1,2. Coupled to lipolysis is the release of a recently identified hormone, fatty-acid-binding protein 4 (FABP4)3. Although circulating FABP4 levels have been strongly associated with cardiometabolic diseases in both preclinical models and humans4-7, no mechanism of action has yet been described8-10. Here we show that hormonal FABP4 forms a functional hormone complex with adenosine kinase (ADK) and nucleoside diphosphate kinase (NDPK) to regulate extracellular ATP and ADP levels. We identify a substantial effect of this hormone on beta cells and given the central role of beta-cell function in both the control of lipolysis and development of diabetes, postulate that hormonal FABP4 is a key regulator of an adipose-beta-cell endocrine axis. Antibody-mediated targeting of this hormone complex improves metabolic outcomes, enhances beta-cell function and preserves beta-cell integrity to prevent both type 1 and type 2 diabetes. Thus, the FABP4-ADK-NDPK complex, Fabkin, represents a previously unknown hormone and mechanism of action that integrates energy status with the function of metabolic organs, and represents a promising target against metabolic disease.
Collapse
Affiliation(s)
- Kacey J Prentice
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Jani Saksi
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Lauren T Robertson
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Grace Y Lee
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Karen E Inouye
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Kosei Eguchi
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Alexandra Lee
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Ozgur Cakici
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Emily Otterbeck
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Paulina Cedillo
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Ediz S Calay
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Feyza Engin
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA.,Departments of Biomolecular Chemistry and Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA. .,Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
28
|
CEBPβ binding directly to the promoter region drives CEBPɑ transcription and improves FABP4 transcriptional activity in adipose tissue of yak (Bos grunniens). Res Vet Sci 2021; 141:174-179. [PMID: 34749102 DOI: 10.1016/j.rvsc.2021.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/07/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022]
Abstract
Fatty acid binding protein 4 (FABP4) was crucial to fatty acid uptake and intracellular transport. However, the mechanisms regulating yak (Bos grunniens) FABP4 transcription were not determined. In the current study, predominant expression levels of yak FABP4 were identified in subcutaneous fat and longissimus dorsi muscles by quantitative real-time polymerase chain reactions (qPCR). The CCAAT/enhancer binding protein alpha (CEBPα) and myocyte enhancer factor 2A (MEF2A), as transcriptional activator or repressor in the promoter region of FABP4, were confirmed by both site-directed mutagenesis experiment and chromatin immunoprecipitation assay. Additionally, molecular mechanisms of CEBPɑ regulation were analyzed to explore the transcriptional regulatory property of FABP4, which indicated that transcriptional activity of CEBPɑ depended on CCAAT/ enhancer binding protein beta (CEBPβ) transcription factor. Our results demonstrated that CEBPβ binding directly to the promoter region drove CEBPɑ transcription, improving yak FABP4 transcriptional activity in adipocytes. This mechanism expanded the information on the transcriptional regulatory network of adipogenesis.
Collapse
|
29
|
Espinoza C, Fuenzalida B, Leiva A. Increased Fetal Cardiovascular Disease Risk: Potential Synergy Between Gestational Diabetes Mellitus and Maternal Hypercholesterolemia. Curr Vasc Pharmacol 2021; 19:601-623. [PMID: 33902412 DOI: 10.2174/1570161119666210423085407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 03/16/2021] [Indexed: 01/25/2023]
Abstract
Cardiovascular diseases (CVD) remain a major cause of death worldwide. Evidence suggests that the risk for CVD can increase at the fetal stages due to maternal metabolic diseases, such as gestational diabetes mellitus (GDM) and maternal supraphysiological hypercholesterolemia (MSPH). GDM is a hyperglycemic, inflammatory, and insulin-resistant state that increases plasma levels of free fatty acids and triglycerides, impairs endothelial vascular tone regulation, and due to the increased nutrient transport, exposes the fetus to the altered metabolic conditions of the mother. MSPH involves increased levels of cholesterol (mainly as low-density lipoprotein cholesterol) which also causes endothelial dysfunction and alters nutrient transport to the fetus. Despite that an association has already been established between MSPH and increased CVD risk, however, little is known about the cellular processes underlying this relationship. Our knowledge is further obscured when the simultaneous presentation of MSPH and GDM takes place. In this context, GDM and MSPH may substantially increase fetal CVD risk due to synergistic impairment of placental nutrient transport and endothelial dysfunction. More studies on the separate and/or cumulative role of both processes are warranted to suggest specific treatment options.
Collapse
Affiliation(s)
- Cristian Espinoza
- Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - Andrea Leiva
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Providencia 7510157, Chile
| |
Collapse
|
30
|
Harvey I, Stephens JM. Artemisia scoparia promotes adipogenesis in the absence of adipogenic effectors. Obesity (Silver Spring) 2021; 29:1309-1319. [PMID: 34227239 PMCID: PMC8883808 DOI: 10.1002/oby.23199] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Extracts of Artemisia scoparia (SCO) have antidiabetic properties in mice and enhance adipogenesis in vitro, but the underlying mechanisms are unknown. Thiazolidinediones, including rosiglitazone (ROSI), are pharmacological activators of peroxisome proliferator-activated receptor gamma that also promote adipogenesis. The aim of this study was to examine adipogenic pathways responsible for SCO-mediated adipogenesis and identify potential differences between SCO and ROSI in the ability to promote adipocyte development. METHODS The ability of SCO or ROSI to promote adipogenesis in 3T3-L1 cells following systematic omission of the common triad of adipogenic effectors dexamethasone, 1-methyl-3-isobutylxanthine (MIX), and insulin was examined. Adipogenesis was assessed by both neutral lipid quantitation and adipocyte marker gene expression. RESULTS The results demonstrate that SCO and ROSI promote adipogenesis and increase the expression of several peroxisome proliferator-activated receptor gamma target genes involved in lipid accumulation in the absence of MIX. However, ROSI can enhance adipogenesis in the absence of MIX and insulin and differentially regulates adipogenic and lipid metabolism genes as compared with SCO. CONCLUSIONS These data demonstrate the adipogenic capabilities of SCO are similar but not identical to ROSI, thereby warranting further research into SCO as a promising source of therapeutic compounds in the treatment of metabolic disease states.
Collapse
Affiliation(s)
| | - Jacqueline M. Stephens
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803
- To whom correspondence should be addressed Jacqueline Stephens, Louisiana State University, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, Phone (225) 763-2648, FAX (225) 578-2597,
| |
Collapse
|
31
|
Strutt B, Szlapinski S, Gnaneswaran T, Donegan S, Hill J, Bennett J, Hill DJ. Ontology of the apelinergic system in mouse pancreas during pregnancy and relationship with β-cell mass. Sci Rep 2021; 11:15475. [PMID: 34326390 PMCID: PMC8322410 DOI: 10.1038/s41598-021-94725-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023] Open
Abstract
The apelin receptor (Aplnr) and its ligands, Apelin and Apela, contribute to metabolic control. The insulin resistance associated with pregnancy is accommodated by an expansion of pancreatic β-cell mass (BCM) and increased insulin secretion, involving the proliferation of insulin-expressing, glucose transporter 2-low (Ins+Glut2LO) progenitor cells. We examined changes in the apelinergic system during normal mouse pregnancy and in pregnancies complicated by glucose intolerance with reduced BCM. Expression of Aplnr, Apelin and Apela was quantified in Ins+Glut2LO cells isolated from mouse pancreata and found to be significantly higher than in mature β-cells by DNA microarray and qPCR. Apelin was localized to most β-cells by immunohistochemistry although Aplnr was predominantly associated with Ins+Glut2LO cells. Aplnr-staining cells increased three- to four-fold during pregnancy being maximal at gestational days (GD) 9-12 but were significantly reduced in glucose intolerant mice. Apelin-13 increased β-cell proliferation in isolated mouse islets and INS1E cells, but not glucose-stimulated insulin secretion. Glucose intolerant pregnant mice had significantly elevated serum Apelin levels at GD 9 associated with an increased presence of placental IL-6. Placental expression of the apelinergic axis remained unaltered, however. Results show that the apelinergic system is highly expressed in pancreatic β-cell progenitors and may contribute to β-cell proliferation in pregnancy.
Collapse
Affiliation(s)
- Brenda Strutt
- Lawson Health Research Institute, St Joseph Health Care, 268 Grosvenor St, London, ON, N6A 4V2, Canada
| | - Sandra Szlapinski
- Lawson Health Research Institute, St Joseph Health Care, 268 Grosvenor St, London, ON, N6A 4V2, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, N6A 3K7, Canada
| | - Thineesha Gnaneswaran
- Department of Physiology and Pharmacology, Western University, London, ON, N6A 3K7, Canada
| | - Sarah Donegan
- Department of Physiology and Pharmacology, Western University, London, ON, N6A 3K7, Canada
| | - Jessica Hill
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Jamie Bennett
- Lawson Health Research Institute, St Joseph Health Care, 268 Grosvenor St, London, ON, N6A 4V2, Canada
- Life Sciences Program, School of Interdisciplinary Science, McMaster University, Hamilton, ON, L8S 4LD, Canada
| | - David J Hill
- Lawson Health Research Institute, St Joseph Health Care, 268 Grosvenor St, London, ON, N6A 4V2, Canada.
- Department of Physiology and Pharmacology, Western University, London, ON, N6A 3K7, Canada.
- Departments of Medicine and Paediatrics, Western University, London, ON, N6A 3K7, Canada.
| |
Collapse
|
32
|
He YL, Chen MT, Wang T, Zhang MM, Li YX, Wang HY, Ding N. Development of FABP4/5 inhibitors with potential therapeutic effect on type 2 Diabetes Mellitus. Eur J Med Chem 2021; 224:113720. [PMID: 34332399 DOI: 10.1016/j.ejmech.2021.113720] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/11/2021] [Accepted: 07/23/2021] [Indexed: 01/04/2023]
Abstract
Fatty acid-binding protein 4 (FABP4) and fatty acid-binding protein 5 (FABP5) are promising therapeutic targets for the treatment of various metabolic diseases. However, the weak potency, low selectivity over FABP3, or poor pharmacokinetic profiles of currently reported dual FABP4/5 inhibitors impeded further research. Here, we described the characterization of a series of dual FABP4/5 inhibitors with improved metabolic stabilities and physicochemical properties based on our previous studies. Among the compounds, D9 and E1 exhibited good inhibitory activities against FABP4/5 and favorable selectivity over FABP3 in vitro. In cell-based assays, D9 and E1 exerted a decrease of FABP4 secretion, a strong anti-lipolytic effect in mature adipocytes, and suppression of MCP-1 expression in THP-1 macrophages. Moreover, D9 and E1 possessed good metabolic stabilities in mouse hepatic microsomes and acceptable pharmacokinetics profiles in ICR mice. Further in vivo experiments showed that D9 and E1 could potently decrease serum FABP4 levels and ameliorate glucose metabolism disorders in obese diabetic db/db mice. These results demonstrated that D9 and E1 could serve as lead compounds for the development of novel anti-diabetic drugs.
Collapse
Affiliation(s)
- Yu-Long He
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Meng-Ting Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ting Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ming-Ming Zhang
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Ying-Xia Li
- School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - He-Yao Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Ning Ding
- School of Pharmacy, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
33
|
Wang H, Cao J, Su JB, Wang XQ, Wang X, Zhang DM, Wang XH. Serum fatty acid-binding protein 4 levels and responses of pancreatic islet β-cells and α-cells in patients with type 2 diabetes. Diabetol Metab Syndr 2021; 13:70. [PMID: 34174950 PMCID: PMC8234651 DOI: 10.1186/s13098-021-00690-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Serum fatty acid-binding protein 4 (FABP4), as an intracellular lipid chaperone and adipokine, was reported to be related to the incidence of type 2 diabetes (T2D) and diabetic complications, but its association with pancreatic islet β-cell and α-cell functions has not been fully elucidated. So the present study was to investigate the serum FABP4 levels and responses of islet β-cells and α-cells in patients with T2D. METHODS 115 patients with T2D and 89 healthy controls (HC), who received serum FABP4 levels test, were recruited to participate in this study. Moreover, 75-g oral glucose tolerance test (OGTT) was performed in T2D patients to evaluate islet β-cell and α-cell functions. Systemic insulin sensitivity and overall insulin secretion of islet β-cell function were assessed by Matsuda index using C peptide (ISIM-cp) and ratio of the area under the C peptide curve to the glucose curve (AUCcp/glu) during OGTT, respectively. Fasting glucagon (Gluca0min) and postchallenge glucagon assessed by the area under the glucagon curve (AUCgluca) were determined during OGTT to evaluate islet α-cell function. And other various clinical variables were also measured in all participants. Skewed variables were natural log-transformed (ln), such as lnFABP4. RESULTS The serum FABP4 levels in T2D patients were significantly higher than those in HC (p < 0.05). And after partially adjusting for fasting plasma glucose, serum lnFABP4 levels were negatively correlated with lnISIM-cp (r = - 0.332, p < 0.001) and positively correlated with lnAUCcp/glu (r = 0.324, p < 0.001), lnGluca0min (r = 0.200, p = 0.040) and lnAUCgluca (r = 0.311, p < 0.001), respectively, in patients with T2D. Furthermore, when multiple linear regression analyses were applied to adjust for other various clinical variables, serum lnFABP4 levels were found to remain associated with lnISIM-cp (β = - 0.296, t = - 2.900, p = 0.005), lnAUCcp/glu (β = 0.223, t = 2.038, p = 0.046), lnGluca0min (β = 0.272, t = 2.330, p = 0.024) and lnAUCgluca (β = 0.341, t = 3.065, p = 0.004), respectively. CONCLUSION Increased serum FABP4 levels were closely associated with blunted insulin sensitivity, increased insulin secretion, and elevated fasting and postchallenge glucagon levels in patients with T2D.
Collapse
Affiliation(s)
- Hong Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Jie Cao
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Jian-bin Su
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Xue-qin Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Xing Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Dong-mei Zhang
- Medical Research Center, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Xiao-hua Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| |
Collapse
|
34
|
Molinari F, Feraco A, Mirabilii S, Saladini S, Sansone L, Vernucci E, Tomaselli G, Marzolla V, Rotili D, Russo MA, Ricciardi MR, Tafuri A, Mai A, Caprio M, Tafani M, Armani A. SIRT5 Inhibition Induces Brown Fat-Like Phenotype in 3T3-L1 Preadipocytes. Cells 2021; 10:cells10051126. [PMID: 34066961 PMCID: PMC8148511 DOI: 10.3390/cells10051126] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022] Open
Abstract
Brown adipose tissue (BAT) activity plays a key role in regulating systemic energy. The activation of BAT results in increased energy expenditure, making this tissue an attractive pharmacological target for therapies against obesity and type 2 diabetes. Sirtuin 5 (SIRT5) affects BAT function by regulating adipogenic transcription factor expression and mitochondrial respiration. We analyzed the expression of SIRT5 in the different adipose depots of mice. We treated 3T3-L1 preadipocytes and mouse primary preadipocyte cultures with the SIRT5 inhibitor MC3482 and investigated the effects of this compound on adipose differentiation and function. The administration of MC3482 during the early stages of differentiation promoted the expression of brown adipocyte and mitochondrial biogenesis markers. Upon treatment with MC3482, 3T3-L1 adipocytes showed an increased activation of the AMP-activated protein kinase (AMPK), which is known to stimulate brown adipocyte differentiation. This effect was paralleled by an increase in autophagic/mitophagic flux and a reduction in lipid droplet size, mediated by a higher lipolytic rate. Of note, MC3482 increased the expression and the activity of adipose triglyceride lipase, without modulating hormone-sensitive lipase. Our findings reveal that SIRT5 inhibition stimulates brown adipogenesis in vitro, supporting this approach as a strategy to stimulate BAT and counteract obesity.
Collapse
Affiliation(s)
- Francesca Molinari
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (F.M.); (S.S.); (M.T.)
| | - Alessandra Feraco
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00163 Rome, Italy; (A.F.); (V.M.); (M.C.)
| | - Simone Mirabilii
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
| | - Serena Saladini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (F.M.); (S.S.); (M.T.)
| | - Luigi Sansone
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele, 00166 Rome, Italy; (L.S.); (G.T.); (M.A.R.)
| | - Enza Vernucci
- Department of Cardiovascular, Nephrologic, Anesthesiologic and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy;
| | - Giada Tomaselli
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele, 00166 Rome, Italy; (L.S.); (G.T.); (M.A.R.)
| | - Vincenzo Marzolla
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00163 Rome, Italy; (A.F.); (V.M.); (M.C.)
| | - Dante Rotili
- Department of Chemistry and Technology of Drugs, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy; (D.R.); (A.M.)
| | - Matteo A. Russo
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele, 00166 Rome, Italy; (L.S.); (G.T.); (M.A.R.)
- MEBIC Consortium, San Raffaele Rome Open University, 00166 Rome, Italy
| | - Maria Rosaria Ricciardi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
| | - Agostino Tafuri
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
- Hematology, “Sant’ Andrea” University Hospital, Sapienza University of Rome, 00189 Rome, Italy
| | - Antonello Mai
- Department of Chemistry and Technology of Drugs, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy; (D.R.); (A.M.)
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00163 Rome, Italy; (A.F.); (V.M.); (M.C.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (F.M.); (S.S.); (M.T.)
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00163 Rome, Italy; (A.F.); (V.M.); (M.C.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Correspondence:
| |
Collapse
|
35
|
Droujinine IA, Meyer AS, Wang D, Udeshi ND, Hu Y, Rocco D, McMahon JA, Yang R, Guo J, Mu L, Carey DK, Svinkina T, Zeng R, Branon T, Tabatabai A, Bosch JA, Asara JM, Ting AY, Carr SA, McMahon AP, Perrimon N. Proteomics of protein trafficking by in vivo tissue-specific labeling. Nat Commun 2021; 12:2382. [PMID: 33888706 PMCID: PMC8062696 DOI: 10.1038/s41467-021-22599-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Conventional approaches to identify secreted factors that regulate homeostasis are limited in their abilities to identify the tissues/cells of origin and destination. We established a platform to identify secreted protein trafficking between organs using an engineered biotin ligase (BirA*G3) that biotinylates, promiscuously, proteins in a subcellular compartment of one tissue. Subsequently, biotinylated proteins are affinity-enriched and identified from distal organs using quantitative mass spectrometry. Applying this approach in Drosophila, we identify 51 muscle-secreted proteins from heads and 269 fat body-secreted proteins from legs/muscles, including CG2145 (human ortholog ENDOU) that binds directly to muscles and promotes activity. In addition, in mice, we identify 291 serum proteins secreted from conditional BirA*G3 embryo stem cell-derived teratomas, including low-abundance proteins with hormonal properties. Our findings indicate that the communication network of secreted proteins is vast. This approach has broad potential across different model systems to identify cell-specific secretomes and mediators of interorgan communication in health or disease.
Collapse
Affiliation(s)
- Ilia A Droujinine
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA.
| | - Amanda S Meyer
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Dan Wang
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Entomology, China Agricultural University, Beijing, China
| | | | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - David Rocco
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Rui Yang
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - JinJin Guo
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Luye Mu
- Department of Electrical Engineering, Yale University, New Haven, CT, USA
| | | | | | - Rebecca Zeng
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Tess Branon
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Areya Tabatabai
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Justin A Bosch
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - John M Asara
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Alice Y Ting
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Departments of Genetics, Biology, and Chemistry, Stanford University, Stanford, CA, USA
| | - Steven A Carr
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
36
|
Lee CH, Lui DTW, Lam KSL. Adipocyte Fatty Acid-Binding Protein, Cardiovascular Diseases and Mortality. Front Immunol 2021; 12:589206. [PMID: 33815359 PMCID: PMC8017191 DOI: 10.3389/fimmu.2021.589206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 03/04/2021] [Indexed: 12/21/2022] Open
Abstract
It has been increasingly recognized that inflammation plays an important role in the pathogenesis of cardiovascular disease (CVD). In obesity, adipose tissue inflammation, especially in the visceral fat depots, contributes to systemic inflammation and promotes the development of atherosclerosis. Adipocyte fatty acid-binding protein (AFABP), a lipid chaperone abundantly secreted from the adipocytes and macrophages, is one of the key players mediating this adipose-vascular cross-talk, in part via its interaction with c-Jun NH2-terminal kinase (JNK) and activator protein-1 (AP-1) to form a positive feedback loop, and perpetuate inflammatory responses. In mice, selective JNK inactivation in the adipose tissue significantly reduced the expression of AFABP in their adipose tissue, as well as circulating AFABP levels. Importantly, fat transplant experiments showed that adipose-specific JNK inactivation in the visceral fat was sufficient to protect mice with apoE deficiency from atherosclerosis, with the beneficial effects attenuated by the continuous infusion of recombinant AFABP, supporting the role of AFABP as the link between visceral fat inflammation and atherosclerosis. In humans, raised circulating AFABP levels are associated with incident metabolic syndrome, type 2 diabetes and CVD, as well as non-alcoholic steatohepatitis, diabetic nephropathy and adverse renal outcomes, all being conditions closely related to inflammation and enhanced CV mortality. Collectively, these clinical data have provided support to AFABP as an important adipokine linking obesity, inflammation and CVD. This review will discuss recent findings on the role of AFABP in CVD and mortality, the possible underlying mechanisms, and pharmacological inhibition of AFABP as a potential strategy to combat CVD.
Collapse
Affiliation(s)
- Chi-Ho Lee
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, Hong Kong
| | - David T W Lui
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| | - Karen S L Lam
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
37
|
Papathanasiou AE, Spyropoulos F, Michael Z, Joung KE, Briana DD, Malamitsi-Puchner A, Mantzoros CS, Christou H. Adipokines and Metabolic Regulators in Human and Experimental Pulmonary Arterial Hypertension. Int J Mol Sci 2021; 22:ijms22031435. [PMID: 33535425 PMCID: PMC7867052 DOI: 10.3390/ijms22031435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Pulmonary hypertension (PH) is associated with meta-inflammation related to obesity but the role of adipose tissue in PH pathogenesis is unknown. We hypothesized that adipose tissue-derived metabolic regulators are altered in human and experimental PH. We measured circulating levels of fatty acid binding protein 4 (FABP-4), fibroblast growth factor -21 (FGF-21), adiponectin, and the mRNA levels of FABP-4, FGF-21, and peroxisome proliferator-activated receptor γ (PPARγ) in lung tissue of patients with idiopathic PH and healthy controls. We also evaluated lung and adipose tissue expression of these mediators in the three most commonly used experimental rodent models of pulmonary hypertension. Circulating levels of FABP-4, FGF-21, and adiponectin were significantly elevated in PH patients compared to controls and the mRNA levels of these regulators and PPARγ were also significantly increased in human PH lungs and in the lungs of rats with experimental PH compared to controls. These findings were coupled with increased levels of adipose tissue mRNA of genes related to glucose uptake, glycolysis, tricarboxylic acid cycle, and fatty acid oxidation in experimental PH. Our results support that metabolic alterations in human PH are recapitulated in rodent models of the disease and suggest that adipose tissue may contribute to PH pathogenesis.
Collapse
Affiliation(s)
- Aimilia Eirini Papathanasiou
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (A.E.P.); (F.S.); (K.E.J.)
- Harvard Medical School, Boston, MA 02215, USA;
- Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 10679 Athens, Greece; (D.D.B.); (A.M.-P.)
| | - Fotios Spyropoulos
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (A.E.P.); (F.S.); (K.E.J.)
- Harvard Medical School, Boston, MA 02215, USA;
| | - Zoe Michael
- Harvard Medical School, Boston, MA 02215, USA;
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02215, USA
| | - Kyoung E. Joung
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (A.E.P.); (F.S.); (K.E.J.)
- Harvard Medical School, Boston, MA 02215, USA;
| | - Despina D. Briana
- Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 10679 Athens, Greece; (D.D.B.); (A.M.-P.)
| | - Ariadne Malamitsi-Puchner
- Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 10679 Athens, Greece; (D.D.B.); (A.M.-P.)
| | - Christos S. Mantzoros
- Harvard Medical School, Boston, MA 02215, USA;
- Division of Endocrinology Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02215, USA
- Correspondence: (C.S.M.); (H.C.)
| | - Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (A.E.P.); (F.S.); (K.E.J.)
- Harvard Medical School, Boston, MA 02215, USA;
- Correspondence: (C.S.M.); (H.C.)
| |
Collapse
|
38
|
Morales LD, Cromack DT, Tripathy D, Fourcaudot M, Kumar S, Curran JE, Carless M, Göring HHH, Hu SL, Lopez-Alvarenga JC, Garske KM, Pajukanta P, Small KS, Glastonbury CA, Das SK, Langefeld C, Hanson RL, Hsueh WC, Norton L, Arya R, Mummidi S, Blangero J, DeFronzo RA, Duggirala R, Jenkinson CP. Further evidence supporting a potential role for ADH1B in obesity. Sci Rep 2021; 11:1932. [PMID: 33479282 PMCID: PMC7820614 DOI: 10.1038/s41598-020-80563-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 12/06/2020] [Indexed: 01/22/2023] Open
Abstract
Insulin is an essential hormone that regulates glucose homeostasis and metabolism. Insulin resistance (IR) arises when tissues fail to respond to insulin, and it leads to serious health problems including Type 2 Diabetes (T2D). Obesity is a major contributor to the development of IR and T2D. We previously showed that gene expression of alcohol dehydrogenase 1B (ADH1B) was inversely correlated with obesity and IR in subcutaneous adipose tissue of Mexican Americans. In the current study, a meta-analysis of the relationship between ADH1B expression and BMI in Mexican Americans, African Americans, Europeans, and Pima Indians verified that BMI was increased with decreased ADH1B expression. Using established human subcutaneous pre-adipocyte cell lines derived from lean (BMI < 30 kg m-2) or obese (BMI ≥ 30 kg m-2) donors, we found that ADH1B protein expression increased substantially during differentiation, and overexpression of ADH1B inhibited fatty acid binding protein expression. Mature adipocytes from lean donors expressed ADH1B at higher levels than obese donors. Insulin further induced ADH1B protein expression as well as enzyme activity. Knockdown of ADH1B expression decreased insulin-stimulated glucose uptake. Our findings suggest that ADH1B is involved in the proper development and metabolic activity of adipose tissues and this function is suppressed by obesity.
Collapse
Affiliation(s)
- Liza D Morales
- South Texas Diabetes and Obesity Institute Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, TX, USA.
| | | | - Devjit Tripathy
- South Texas Veterans Health Care System, San Antonio, TX, USA
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Marcel Fourcaudot
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Satish Kumar
- South Texas Diabetes and Obesity Institute Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, TX, USA
| | - Joanne E Curran
- South Texas Diabetes and Obesity Institute Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, TX, USA
| | - Melanie Carless
- Department of Population Health, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Harald H H Göring
- South Texas Diabetes and Obesity Institute Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, TX, USA
| | - Shirley L Hu
- University of Texas Health Houston, School of Public Health, Brownsville, TX, USA
| | - Juan Carlos Lopez-Alvarenga
- South Texas Diabetes and Obesity Institute Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, TX, USA
| | - Kristina M Garske
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | | | - Swapan K Das
- Internal Medicine-Endocrinology and Metabolism, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Carl Langefeld
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Robert L Hanson
- Phoenix Epidemiology and Clinical Research Branch, NIDDK, Phoenix, AZ, USA
| | - Wen-Chi Hsueh
- Phoenix Epidemiology and Clinical Research Branch, NIDDK, Phoenix, AZ, USA
| | - Luke Norton
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Rector Arya
- South Texas Diabetes and Obesity Institute Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, TX, USA
| | - Srinivas Mummidi
- South Texas Diabetes and Obesity Institute Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, TX, USA
| | - John Blangero
- South Texas Diabetes and Obesity Institute Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, TX, USA
| | - Ralph A DeFronzo
- South Texas Veterans Health Care System, San Antonio, TX, USA
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Ravindranath Duggirala
- South Texas Diabetes and Obesity Institute Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, TX, USA
| | - Christopher P Jenkinson
- South Texas Diabetes and Obesity Institute Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, TX, USA.
| |
Collapse
|
39
|
Nguyen-Tu MS, Martinez-Sanchez A, Leclerc I, Rutter GA, da Silva Xavier G. Adipocyte-specific deletion of Tcf7l2 induces dysregulated lipid metabolism and impairs glucose tolerance in mice. Diabetologia 2021; 64:129-141. [PMID: 33068125 PMCID: PMC7567653 DOI: 10.1007/s00125-020-05292-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Transcription factor 7-like 2 (TCF7L2) is a downstream effector of the Wnt/β-catenin signalling pathway implicated in type 2 diabetes risk through genome-wide association studies. Although its expression is critical for adipocyte development, the potential roles of changes in adipose tissue TCF7L2 levels in diabetes risk are poorly defined. Here, we investigated whether forced changes in Tcf7l2 expression in adipocytes affect whole body glucose or lipid metabolism and crosstalk between disease-relevant tissues. METHODS Tcf7l2 was selectively ablated in mature adipocytes in C57BL/6J mice using Cre recombinase under Adipoq promoter control to recombine Tcf7l2 alleles floxed at exon 1 (referred to as aTCF7L2 mice). aTCF7L2 mice were fed normal chow or a high-fat diet for 12 weeks. Glucose and insulin sensitivity, as well as beta cell function, were assessed in vivo and in vitro. Levels of circulating NEFA, selected hormones and adipokines were measured using standard assays. RESULTS Reduced TCF7L2 expression in adipocytes altered glucose tolerance and insulin secretion in male but not in female mice. Thus, on a normal chow diet, male heterozygote knockout mice (aTCF7L2het) exhibited impaired glucose tolerance at 16 weeks (p = 0.03) and increased fat mass (1.4 ± 0.1-fold, p = 0.007) but no changes in insulin secretion. In contrast, male homozygote knockout (aTCF7L2hom) mice displayed normal body weight but impaired oral glucose tolerance at 16 weeks (p = 0.0001). These changes were mechanistically associated with impaired in vitro glucose-stimulated insulin secretion (decreased 0.5 ± 0.1-fold vs control mice, p = 0.02) and decreased levels of the incretins glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide (0.6 ± 0.1-fold and 0.4 ± 0.1-fold vs control mice, p = 0.04 and p < 0.0001, respectively). Circulating levels of plasma NEFA and fatty acid binding protein 4 were increased by 1.3 ± 0.1-fold and 1.8 ± 0.3-fold vs control mice (p = 0.03 and p = 0.05, respectively). Following exposure to a high-fat diet for 12 weeks, male aTCF7L2hom mice exhibited reduced in vivo glucose-stimulated insulin secretion (0.5 ± 0.1-fold vs control mice, p = 0.02). CONCLUSIONS/INTERPRETATION Loss of Tcf7l2 gene expression selectively in adipocytes leads to a sexually dimorphic phenotype, with impairments not only in adipocytes, but also in pancreatic islet and enteroendocrine cells in male mice only. Our findings suggest novel roles for adipokines and incretins in the effects of diabetes-associated variants in TCF7L2, and further illuminate the roles of TCF7L2 in glucose homeostasis and diabetes risk. Graphical abstract.
Collapse
Affiliation(s)
- Marie-Sophie Nguyen-Tu
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Hammersmith Hospital, Imperial College Centre for Translational and Experimental Medicine, London, UK
| | - Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Hammersmith Hospital, Imperial College Centre for Translational and Experimental Medicine, London, UK
| | - Isabelle Leclerc
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Hammersmith Hospital, Imperial College Centre for Translational and Experimental Medicine, London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Hammersmith Hospital, Imperial College Centre for Translational and Experimental Medicine, London, UK.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | - Gabriela da Silva Xavier
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Hammersmith Hospital, Imperial College Centre for Translational and Experimental Medicine, London, UK.
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK.
| |
Collapse
|
40
|
Wang Z, Guo W, Yi F, Zhou T, Li X, Feng Y, Guo Q, Xu H, Song X, Cao L. The Regulatory Effect of SIRT1 on Extracellular Microenvironment Remodeling. Int J Biol Sci 2021; 17:89-96. [PMID: 33390835 PMCID: PMC7757024 DOI: 10.7150/ijbs.52619] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
The sirtuins family is well known by its unique nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase function. The most-investigated member of the family, Sirtuin 1 (SIRT1), accounts for deacetylating a broad range of transcription factors and coregulators, such as p53, the Forkhead box O (FOXO), and so on. It serves as a pivotal regulator in various intracellular biological processes, including energy metabolism, DNA damage response, genome stability maintenance and tumorigenesis. Although the most attention has been focused on its intracellular functions, the regulatory effect on extracellular microenvironment remodeling of SIRT1 has been recognized by researchers recently. SIRT1 can regulate cell secretion process and participate in glucose metabolism, neuroendocrine function, inflammation and tumorigenesis. Here, we review the advances in the understanding of SIRT1 on remodeling the extracellular microenvironment, which may provide new ideas for pathogenesis investigation and guidance for clinical treatment.
Collapse
Affiliation(s)
- Zhuo Wang
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Wendong Guo
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Fei Yi
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Tingting Zhou
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Xiaoman Li
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Yanling Feng
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Qiqiang Guo
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Hongde Xu
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Xiaoyu Song
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Liu Cao
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| |
Collapse
|
41
|
D’Anneo A, Bavisotto CC, Gammazza AM, Paladino L, Carlisi D, Cappello F, de Macario EC, Macario AJL, Lauricella M. Lipid chaperones and associated diseases: a group of chaperonopathies defining a new nosological entity with implications for medical research and practice. Cell Stress Chaperones 2020; 25:805-820. [PMID: 32856199 PMCID: PMC7591661 DOI: 10.1007/s12192-020-01153-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/16/2020] [Accepted: 08/10/2020] [Indexed: 02/08/2023] Open
Abstract
Fatty acid-binding proteins (FABPs) are lipid chaperones assisting in the trafficking of long-chain fatty acids with functions in various cell compartments, including oxidation, signaling, gene-transcription regulation, and storage. The various known FABP isoforms display distinctive tissue distribution, but some are active in more than one tissue. Quantitative and/or qualitative changes of FABPs are associated with pathological conditions. Increased circulating levels of FABPs are biomarkers of disorders such as obesity, insulin resistance, cardiovascular disease, and cancer. Deregulated expression and malfunction of FABPs can result from genetic alterations or posttranslational modifications and can be pathogenic. We have assembled the disorders with abnormal FABPs as chaperonopathies in a distinct nosological entity. This entity is similar but separate from that encompassing the chaperonopathies pertaining to protein chaperones. In this review, we discuss the role of FABPs in the pathogenesis of metabolic syndrome, cancer, and neurological diseases. We highlight the opportunities for improving diagnosis and treatment that open by encompassing all these pathological conditions within of a coherent nosological group, focusing on abnormal lipid chaperones as biomarkers of disease and etiological-pathogenic factors.
Collapse
Affiliation(s)
- Antonella D’Anneo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Laboratory of Biochemistry, University of Palermo, 90127 Palermo, Italy
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Anatomy, University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Anatomy, University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Letizia Paladino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Anatomy, University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Daniela Carlisi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy
| | - Francesco Cappello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Anatomy, University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Everly Conway de Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202 USA
| | - Alberto J. L. Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202 USA
| | - Marianna Lauricella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
42
|
Wu KKL, Cheung SWM, Cheng KKY. NLRP3 Inflammasome Activation in Adipose Tissues and Its Implications on Metabolic Diseases. Int J Mol Sci 2020; 21:E4184. [PMID: 32545355 PMCID: PMC7312293 DOI: 10.3390/ijms21114184] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
Adipose tissue is an active endocrine and immune organ that controls systemic immunometabolism via multiple pathways. Diverse immune cell populations reside in adipose tissue, and their composition and immune responses vary with nutritional and environmental conditions. Adipose tissue dysfunction, characterized by sterile low-grade chronic inflammation and excessive immune cell infiltration, is a hallmark of obesity, as well as an important link to cardiometabolic diseases. Amongst the pro-inflammatory factors secreted by the dysfunctional adipose tissue, interleukin (IL)-1β, induced by the NLR family pyrin domain-containing 3 (NLRP3) inflammasome, not only impairs peripheral insulin sensitivity, but it also interferes with the endocrine and immune functions of adipose tissue in a paracrine manner. Human studies indicated that NLRP3 activity in adipose tissues positively correlates with obesity and its metabolic complications, and treatment with the IL-1β antibody improves glycaemia control in type 2 diabetic patients. In mouse models, genetic or pharmacological inhibition of NLRP3 activation pathways or IL-1β prevents adipose tissue dysfunction, including inflammation, fibrosis, defective lipid handling and adipogenesis, which in turn alleviates obesity and its related metabolic disorders. In this review, we summarize both the negative and positive regulators of NLRP3 inflammasome activation, and its pathophysiological consequences on immunometabolism. We also discuss the potential therapeutic approaches to targeting adipose tissue inflammasome for the treatment of obesity and its related metabolic disorders.
Collapse
Affiliation(s)
| | | | - Kenneth King-Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (K.K.-L.W.); (S.W.-M.C.)
| |
Collapse
|
43
|
Adipokines as key players in β cell function and failure. Clin Sci (Lond) 2020; 133:2317-2327. [PMID: 31769478 DOI: 10.1042/cs20190523] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
The growing prevalence of obesity and its related metabolic diseases, mainly Type 2 diabetes (T2D), has increased the interest in adipose tissue (AT) and its role as a principal metabolic orchestrator. Two decades of research have now shown that ATs act as an endocrine organ, secreting soluble factors termed adipocytokines or adipokines. These adipokines play crucial roles in whole-body metabolism with different mechanisms of action largely dependent on the tissue or cell type they are acting on. The pancreatic β cell, a key regulator of glucose metabolism due to its ability to produce and secrete insulin, has been identified as a target for several adipokines. This review will focus on how adipokines affect pancreatic β cell function and their impact on pancreatic β cell survival in disease contexts such as diabetes. Initially, the "classic" adipokines will be discussed, followed by novel secreted adipocyte-specific factors that show therapeutic promise in regulating the adipose-pancreatic β cell axis.
Collapse
|
44
|
Dou HX, Wang T, Su HX, Gao DD, Xu YC, Li YX, Wang HY. Exogenous FABP4 interferes with differentiation, promotes lipolysis and inflammation in adipocytes. Endocrine 2020; 67:587-596. [PMID: 31845180 DOI: 10.1007/s12020-019-02157-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE Fatty acid binding protein 4 (FABP4) has been demonstrated to be secreted from adipocytes in an unconventional pathway associated with lipolysis. Circulating FABP4 is elevated in metabolic disorders and has been shown to affect various peripheral cells such as pancreatic β-cells, hepatocytes and macrophages, but its effects on adipocytes remains unclear. The aim of this study was to investigate the effects of exogenous FABP4 (eFABP4) on adipocyte differentiation and function. METHODS 3T3-L1 pre-adipocytes or mature adipocytes were treated with recombinant FABP4 in the absence or presence of FABP4 inhibitor I-9/p38 MAPK inhibitor SB203580; Meanwhile male C57BL/6J mice were subcutaneously injected twice a day with recombinant FABP4 (0.35 mg/kg) with or without I-9 (50 mg/kg) for 2 weeks. The effects of eFABP4 on differentiation, lipolysis and inflammation were determined by triglyceride measurement or lipolysis assay, western blotting, or RT-qPCR analysis. RESULTS eFABP4 treatment significantly reduced intracellular triglyceride content and decreased expression of adipogenic markers peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer binding protein alpha (C/EBPα), intracellular FABP4, and adiponectin in 3T3-L1 cells. Besides, eFABP4 promoted lipolysis and inflammation in differentiated 3T3-L1 adipocytes as well as in adipose tissue of eFABP4-treated C57BL/6J mice, with elevated gene expression of monocyte chemoattractant protein (MCP)-1, tumor necrosis factor (TNF)-α, and elevated protein expression of adipose triglyceride lipase (ATGL), phosphorylation of hormone-sensitive lipase (HSL) (Ser-660), p38, and nuclear factor-kappa B (NF-κB). The pro-inflammatory and pro-lipolytic effects of eFABP4 could be reversed by SB203580/I-9. CONCLUSIONS These findings indicate that eFABP4 interferes with adipocyte differentiation, induces p38/HSL mediated lipolysis and p38/NF-κB mediated inflammation in adipocytes in vitro and in vivo.
Collapse
Affiliation(s)
- Hui-Xia Dou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Ting Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hai-Xia Su
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Ding-Ding Gao
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Ye-Chun Xu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ying-Xia Li
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - He-Yao Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
45
|
Abstract
The Special Issue “Pathogenetic and Therapeutic Significance of Adipokines in Diabetes” focused on adipokines as shared diagnostic biomarkers and therapeutic targets for both obesity and type 2 diabetes. Experts discussed the pathological role of adipokines in their studies associated with diabetes. It provided new insights into the role of adipokines in diabetes. In this commentary and review, these studies will be summarized and the novel roles of adipokines will be discussed. This will also confirm the role of adipokines as biomarkers for diagnosis and prediction, and as therapeutic targets of diabetes and its related pathogenic phenomena.
Collapse
|
46
|
Papathanasiou AE, Briana DD, Gavrili S, Georgantzi S, Papathoma E, Marmarinos A, Christou C, Voulgaris K, Gourgiotis D, Malamitsi‐Puchner A. Cord blood fatty acid-binding protein-4 levels are upregulated at both ends of the birthweight spectrum. Acta Paediatr 2019; 108:2083-2088. [PMID: 31025416 DOI: 10.1111/apa.14826] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/13/2019] [Accepted: 04/23/2019] [Indexed: 01/17/2023]
Abstract
AIM Fatty acid-binding protein-4 (FABP4) is an adipokine associated with obesity and signs of the metabolic syndrome. We aimed to investigate at birth in term neonates with normal and abnormal intrauterine growth concentrations of FABP4 and associate them with various perinatal parameters. METHODS Serum cord blood FABP4 levels were prospectively determined by ELISA in 80 singleton term appropriate-for-gestational-age (AGA), intrauterine growth-restricted (IUGR) and large-for-gestational-age (LGA) neonates. RESULTS Compared to the AGA group, cord blood FABP4 levels were increased in the IUGR and LGA groups. Additionally, they were higher in early-term than full-term neonates. A significant U-shaped correlation was recorded between serum FABP4 levels and birthweight. A significant negative correlation between cord blood FABP4 and gestational age in the whole study population was noted. CONCLUSION Cord blood FABP4 levels were significantly higher at the extremes of foetal growth at term and negatively correlated with gestational age, being increased in early-term versus full-term neonates. Further longitudinal studies with larger sample sizes are required to elucidate FABP4 implication in foetal growth and its association with future adverse cardiometabolic outcomes in the offspring.
Collapse
Affiliation(s)
| | - Despina D. Briana
- Medical School National and Kapodistrian University of Athens Athens Greece
| | - Stavroula Gavrili
- Neonatal Intensive Care Unit ‘Alexandra’ University and State Maternity Hospital Athens Greece
| | - Sophia Georgantzi
- Neonatal Intensive Care Unit ‘Alexandra’ University and State Maternity Hospital Athens Greece
| | - Evangelia Papathoma
- Neonatal Intensive Care Unit ‘Alexandra’ University and State Maternity Hospital Athens Greece
| | - Antonios Marmarinos
- Laboratory of Clinical Biochemistry‐Molecular Diagnostics 2nd Department of Pediatrics National and Kapodistrian University of Athens Athens Greece
| | | | | | - Dimitrios Gourgiotis
- Laboratory of Clinical Biochemistry‐Molecular Diagnostics 2nd Department of Pediatrics National and Kapodistrian University of Athens Athens Greece
| | | |
Collapse
|
47
|
Lorenzo-Almorós A, Hang T, Peiró C, Soriano-Guillén L, Egido J, Tuñón J, Lorenzo Ó. Predictive and diagnostic biomarkers for gestational diabetes and its associated metabolic and cardiovascular diseases. Cardiovasc Diabetol 2019; 18:140. [PMID: 31666083 PMCID: PMC6820966 DOI: 10.1186/s12933-019-0935-9] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/21/2019] [Indexed: 12/11/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is defined as the presence of high blood glucose levels with the onset, or detected for the first time during pregnancy, as a result of increased insulin resistance. GDM may be induced by dysregulation of pancreatic β-cell function and/or by alteration of secreted gestational hormones and peptides related with glucose homeostasis. It may affect one out of five pregnancies, leading to perinatal morbidity and adverse neonatal outcomes, and high risk of chronic metabolic and cardiovascular injuries in both mother and offspring. Currently, GDM diagnosis is based on evaluation of glucose homeostasis at late stages of pregnancy, but increased age and body-weight, and familiar or previous occurrence of GDM, may conditionate this criteria. In addition, an earlier and more specific detection of GDM with associated metabolic and cardiovascular risk could improve GDM development and outcomes. In this sense, 1st-2nd trimester-released biomarkers found in maternal plasma including adipose tissue-derived factors such as adiponectin, visfatin, omentin-1, fatty acid-binding protein-4 and retinol binding-protein-4 have shown correlations with GDM development. Moreover, placenta-related factors such as sex hormone-binding globulin, afamin, fetuin-A, fibroblast growth factors-21/23, ficolin-3 and follistatin, or specific micro-RNAs may participate in GDM progression and be useful for its recognition. Finally, urine-excreted metabolites such as those related with serotonin system, non-polar amino-acids and ketone bodies, may complete a predictive or early-diagnostic panel of biomarkers for GDM.
Collapse
Affiliation(s)
- A Lorenzo-Almorós
- Renal, Vascular and Diabetes Laboratory, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Av. Reyes Católicos 2, 28040, Madrid, Spain
| | - T Hang
- Renal, Vascular and Diabetes Laboratory, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Av. Reyes Católicos 2, 28040, Madrid, Spain
| | - C Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - L Soriano-Guillén
- Department of Paediatrics, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - J Egido
- Renal, Vascular and Diabetes Laboratory, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Av. Reyes Católicos 2, 28040, Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM) Network, Madrid, Spain
| | - J Tuñón
- Department of Cardiology, Fundación Jiménez Díaz, Madrid, Spain
| | - Ó Lorenzo
- Renal, Vascular and Diabetes Laboratory, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Av. Reyes Católicos 2, 28040, Madrid, Spain.
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM) Network, Madrid, Spain.
| |
Collapse
|
48
|
Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 2019; 60:1648-1684. [PMID: 31209153 PMCID: PMC6795086 DOI: 10.1194/jlr.r094060] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The breakthrough discoveries of leptin and adiponectin more than two decades ago led to a widespread recognition of adipose tissue as an endocrine organ. Many more adipose tissue-secreted signaling mediators (adipokines) have been identified since then, and much has been learned about how adipose tissue communicates with other organs of the body to maintain systemic homeostasis. Beyond proteins, additional factors, such as lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs), released by adipose tissue participate in this process. Here, we review the diverse signaling mediators and mechanisms adipose tissue utilizes to relay information to other organs. We discuss recently identified adipokines (proteins, lipids, and metabolites) and briefly outline the contributions of noncoding RNAs and EVs to the ever-increasing complexities of adipose tissue inter-organ communication. We conclude by reflecting on central aspects of adipokine biology, namely, the contribution of distinct adipose tissue depots and cell types to adipokine secretion, the phenomenon of adipokine resistance, and the capacity of adipose tissue to act both as a source and sink of signaling mediators.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
49
|
Josephrajan A, Hertzel AV, Bohm EK, McBurney MW, Imai SI, Mashek DG, Kim DH, Bernlohr DA. Unconventional Secretion of Adipocyte Fatty Acid Binding Protein 4 Is Mediated By Autophagic Proteins in a Sirtuin-1-Dependent Manner. Diabetes 2019; 68:1767-1777. [PMID: 31171562 PMCID: PMC6702637 DOI: 10.2337/db18-1367] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Fatty acid binding protein 4 (FABP4) is a leaderless lipid carrier protein primarily expressed by adipocytes and macrophages that not only functions intracellularly but is also secreted. The secretion is mediated via unconventional mechanism(s), and in a variety of species, metabolic dysfunction is correlated with elevated circulating FABP4 levels. In diabetic animals, neutralizing antibodies targeting serum FABP4 increase insulin sensitivity and attenuate hepatic glucose output, suggesting the functional importance of circulating FABP4. Using animal and cell-based models, we show that FABP4 is secreted from white, but not brown, adipose tissue in response to lipolytic stimulation in a sirtuin-1 (SIRT1)-dependent manner via a mechanism that requires some, but not all, autophagic components. Silencing of early autophagic genes such as Ulk1/2, Fip200, or Beclin-1 or chemical inhibition of ULK1/2 or VPS34 attenuated secretion, while Atg5 knockdown potentiated FABP4 release. Genetic knockout of Sirt1 diminished secretion, and serum FABP4 levels were undetectable in Sirt1 knockout mice. In addition, blocking SIRT1 by EX527 attenuated secretion while activating SIRT1 by resveratrol-potentiated secretion. These studies suggest that FABP4 secretion from adipocytes is regulated by SIRT1 and requires early autophagic components.
Collapse
Affiliation(s)
- Ajeetha Josephrajan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Ann V Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Ellie K Bohm
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Michael W McBurney
- Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Ottawa, Ontario, Canada
| | - Shin-Ichiro Imai
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Do-Hyung Kim
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
50
|
Wong YK, Cheung CYY, Tang CS, Au KW, Hai JSH, Lee CH, Lau KK, Cheung BMY, Sham PC, Xu A, Lam KSL, Tse HF. Age-Biomarkers-Clinical Risk Factors for Prediction of Cardiovascular Events in Patients With Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2019; 38:2519-2527. [PMID: 30354221 DOI: 10.1161/atvbaha.118.311726] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Objective- In patients with stable coronary artery disease, conventional risk factors provide limited incremental predictive value for cardiovascular events. We sought to investigate whether a panel of cardiometabolic biomarkers alone or combined with conventional risk factors would exhibit incremental value in the prediction of cardiovascular events. Approach and Results- In the discovery cohort, we measured serum adiponectin, A-FABP (adipocyte fatty acid-binding protein), lipocalin-2, FGF (fibroblast growth factor)-19 and 21, plasminogen activator inhibitor-1, and retinol-binding protein-4 in 1166 Chinese coronary artery disease patients. After a median follow-up of 35 months, 170 patients developed new-onset major adverse cardiovascular events (MACE). In the model with age ≥65 years and conventional risk factors, area under the curve for predicting MACE was 0.68. Addition of lipocalin-2 to the age-clinical risk factor model improved predictive accuracy (area under the curve=0.73). Area under the curve further increased to 0.75 when a combination of lipocalin-2, A-FABP, and FGF-19 was added to yield age-biomarkers-clinical risk factor model. The adjusted hazard ratio on MACEs for lipocalin-2, A-FABP, and FGF-19 levels above optimal cutoffs were 2.23 (95% CI, 1.62-3.08), 1.99 (95% CI, 1.43-2.76), and 1.65 (95% CI, 1.15-2.35), respectively. In the validation cohort of 1262 coronary artery disease patients with type 2 diabetes mellitus, the age-biomarkers-clinical risk factor model was confirmed to provide good discrimination and calibration over the conventional risk factor alone for prediction of MACE. Conclusions- A combination of the 3 biomarkers, lipocalin-2, A-FABP, and FGF-19, with clinical risk factors to yield the age-biomarkers-clinical risk factor model provides an optimal and validated prediction of new-onset MACE in patients with stable coronary artery disease.
Collapse
Affiliation(s)
- Yuen-Kwun Wong
- From the Department of Medicine (Y.-K.W., C.Y.Y.C., K.-W.A., J.S.H.H., C.-H.L., K.-K.L., B.M.Y.C., A.X., K.S.L.L., H.-F.T.), the University of Hong Kong, China
| | - Chloe Y Y Cheung
- From the Department of Medicine (Y.-K.W., C.Y.Y.C., K.-W.A., J.S.H.H., C.-H.L., K.-K.L., B.M.Y.C., A.X., K.S.L.L., H.-F.T.), the University of Hong Kong, China
| | - Clara S Tang
- Department of Surgery (C.S.T.), the University of Hong Kong, China
| | - Ka-Wing Au
- From the Department of Medicine (Y.-K.W., C.Y.Y.C., K.-W.A., J.S.H.H., C.-H.L., K.-K.L., B.M.Y.C., A.X., K.S.L.L., H.-F.T.), the University of Hong Kong, China
| | - JoJo S H Hai
- From the Department of Medicine (Y.-K.W., C.Y.Y.C., K.-W.A., J.S.H.H., C.-H.L., K.-K.L., B.M.Y.C., A.X., K.S.L.L., H.-F.T.), the University of Hong Kong, China
| | - Chi-Ho Lee
- From the Department of Medicine (Y.-K.W., C.Y.Y.C., K.-W.A., J.S.H.H., C.-H.L., K.-K.L., B.M.Y.C., A.X., K.S.L.L., H.-F.T.), the University of Hong Kong, China
| | - Kui-Kai Lau
- From the Department of Medicine (Y.-K.W., C.Y.Y.C., K.-W.A., J.S.H.H., C.-H.L., K.-K.L., B.M.Y.C., A.X., K.S.L.L., H.-F.T.), the University of Hong Kong, China
| | - Bernard M Y Cheung
- From the Department of Medicine (Y.-K.W., C.Y.Y.C., K.-W.A., J.S.H.H., C.-H.L., K.-K.L., B.M.Y.C., A.X., K.S.L.L., H.-F.T.), the University of Hong Kong, China
| | - Pak-Chung Sham
- Department of Psychiatry (P.-C.S.), the University of Hong Kong, China.,Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine (P.-C.S.), the University of Hong Kong, China.,State Key Laboratory in Brain and Cognitive Sciences (P.-C.S.), the University of Hong Kong, China
| | - Aimin Xu
- From the Department of Medicine (Y.-K.W., C.Y.Y.C., K.-W.A., J.S.H.H., C.-H.L., K.-K.L., B.M.Y.C., A.X., K.S.L.L., H.-F.T.), the University of Hong Kong, China.,State Key Laboratory of Pharmaceutical Biotechnology (A.X., K.S.L.L.), the University of Hong Kong, China.,Department of Pharmacology & Pharmacy (A.X.), the University of Hong Kong, China
| | - Karen S L Lam
- From the Department of Medicine (Y.-K.W., C.Y.Y.C., K.-W.A., J.S.H.H., C.-H.L., K.-K.L., B.M.Y.C., A.X., K.S.L.L., H.-F.T.), the University of Hong Kong, China.,State Key Laboratory of Pharmaceutical Biotechnology (A.X., K.S.L.L.), the University of Hong Kong, China
| | - Hung-Fat Tse
- From the Department of Medicine (Y.-K.W., C.Y.Y.C., K.-W.A., J.S.H.H., C.-H.L., K.-K.L., B.M.Y.C., A.X., K.S.L.L., H.-F.T.), the University of Hong Kong, China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine (H.-F.T.), the University of Hong Kong, China.,Shenzhen Institutes of Research and Innovation (H.-F.T.), the University of Hong Kong, China.,Department of Medicine, Shenzhen Hong Kong University Hospital, China (H.-F.T.)
| |
Collapse
|