1
|
Cheong LYT, Saipuljumri EN, Loi GWZ, Zeng J, Lo CH. Autolysosomal Dysfunction in Obesity-induced Metabolic Inflammation and Related Disorders. Curr Obes Rep 2025; 14:43. [PMID: 40366502 PMCID: PMC12078456 DOI: 10.1007/s13679-025-00638-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/15/2025]
Abstract
PURPOSE OF REVIEW Obesity is a global health crisis affecting individuals across all age groups, significantly increasing the risk of metabolic disorders such as type 2 diabetes (T2D), metabolic dysfunction-associated fatty liver disease (MAFLD), and cardiovascular diseases. The World Health Organization reported in 2022 that 2.5 billion adults were overweight, with 890 million classified as obese, emphasizing the urgent need for effective interventions. A critical aspect of obesity's pathophysiology is meta-inflammation-a chronic, systemic low-grade inflammatory state driven by excess adipose tissue, which disrupts metabolic homeostasis. This review examines the role of autolysosomal dysfunction in obesity-related metabolic disorders, exploring its impact across multiple metabolic organs and evaluating potential therapeutic strategies that target autophagy and lysosomal function. RECENT FINDINGS Emerging research highlights the importance of autophagy in maintaining cellular homeostasis and metabolic balance. Obesity-induced lysosomal dysfunction impairs the autophagic degradation process, contributing to the accumulation of damaged organelles and toxic aggregates, exacerbating insulin resistance, lipotoxicity, and chronic inflammation. Studies have identified autophagic defects in key metabolic tissues, including adipose tissue, skeletal muscle, liver, pancreas, kidney, heart, and brain, linking autophagy dysregulation to the progression of metabolic diseases. Preclinical investigations suggest that pharmacological and nutritional interventions-such as AMPK activation, caloric restriction mimetics, and lysosomal-targeting compounds-can restore autophagic function and improve metabolic outcomes in obesity models. Autolysosomal dysfunction is a pivotal contributor to obesity-associated metabolic disorders , influencing systemic inflammation and metabolic dysfunction. Restoring autophagy and lysosomal function holds promise as a therapeutic strategy to mitigate obesity-driven pathologies. Future research should focus on translating these findings into clinical applications, optimizing targeted interventions to improve metabolic health and reduce obesity-associated complications.
Collapse
Affiliation(s)
- Lenny Yi Tong Cheong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | | | - Gavin Wen Zhao Loi
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Jialiu Zeng
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA.
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA.
| | - Chih Hung Lo
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA.
- Department of Biology, Syracuse University, Syracuse, NY, 13244, USA.
| |
Collapse
|
2
|
Heikkinen A, Esser VFC, Lee SHT, Lundgren S, Hakkarainen A, Lundbom J, Kuula J, Groop PH, Heinonen S, Villicaña S, Bell JT, Maguolo A, Nilsson E, Ling C, Vaag A, Pajukanta P, Kaprio J, Pietiläinen KH, Li S, Ollikainen M. Twin pair analysis uncovers links between DNA methylation, mitochondrial DNA quantity and obesity. Nat Commun 2025; 16:4374. [PMID: 40355419 PMCID: PMC12069627 DOI: 10.1038/s41467-025-59576-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
Alterations in mitochondrial metabolism in obesity may indicate disrupted communication between mitochondria and nucleus, and DNA methylation may influence this interplay. Here, we leverage data from the Finnish Twin Cohort study subcohort (n = 173; 86 full twin pairs, 1 singleton), including comprehensive measurements of obesity-related outcomes, mitochondrial DNA quantity and nuclear DNA methylation levels in adipose and muscle tissue, to identify one CpG at SH3BP4 significantly associated with mitochondrial DNA quantity in adipose tissue (FDR < 0.05). We also show that SH3BP4 methylation correlates with its gene expression. Additionally, we find that 14 out of the 35 obesity-related traits display significant associations with both SH3BP4 methylation and mitochondrial DNA quantity in adipose tissue. We use data from TwinsUK and the Scandinavian T2D-discordant monozygotic twin cohort, to validate the observed associations. Further analysis using ICE FALCON suggests that mitochondrial DNA quantity, insulin sensitivity and certain body fat measures are causal to SH3BP4 methylation. Examining mitochondrial DNA quantity and obesity-related traits suggests causation from mitochondrial DNA quantity to obesity, but unmeasured within-individual confounding cannot be ruled out. Our findings underscore the impact of mitochondrial DNA quantity on DNA methylation and expression of the SH3BP4 gene within adipose tissue, with potential implications for obesity.
Collapse
Affiliation(s)
- Aino Heikkinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| | - Vivienne F C Esser
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
| | - Seung Hyuk T Lee
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sara Lundgren
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Antti Hakkarainen
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jesper Lundbom
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany
| | - Juho Kuula
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Sergio Villicaña
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Alice Maguolo
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Emma Nilsson
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Allan Vaag
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
- Copenhagen University Hospital, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Endocrinology, Skåne University Hospital, Malmö, Sweden
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Shuai Li
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| |
Collapse
|
3
|
Hanttu AM, Muniandy M, van der Kolk BW, Lindgren H, Yadav B, Sutinen J, Pietiläinen KH. Adipose tissue transcriptome in patients switching efavirenz or a protease inhibitor to raltegravir compared to people without HIV. AIDS 2025; 39:676-682. [PMID: 39874113 DOI: 10.1097/qad.0000000000004131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
OBJECTIVE The aim of this article is to study the subcutaneous adipose tissue (SAT) transcriptome in people with HIV (PWH) switching efavirenz (EFV) or a protease inhibitor to raltegravir and to compare the transcriptome of PWH to those of people without HIV (PWoH). DESIGN PWH ( n = 36) on EFV ( n = 22) or a protease inhibitor ( n = 14) based ART regimen were randomized to switch to RAL ( n = 15) or to continue unchanged medication ( n = 17). PWoH ( n = 10), comparable in age and BMI, were included for comparison. METHODS SAT gene expression was analyzed via RNA sequencing (Illumina Stranded mRNA library prep). RESULTS At baseline, only 51 out of 19 930 genes showed differential expression (FDR <0.05) between PWH and PWoH. Differentially expressed genes in PWH were identified as being HIV host factors or were associated with immune response, lipid metabolism, adipogenesis, apoptosis regulation, DNA/RNA metabolism, and cell structures. Mitochondria-encoded genes were consistently downregulated in PWH. Intergroup variations among PWH using different ART (EFV, protease inhibitor, RAL) were not significant, and switching EFV or a protease inhibitor to RAL did not induce substantial changes in the SAT transcriptome. CONCLUSION While some specific genes linked to HIV are differentially expressed in PWH compared to PWoH, the overall SAT transcriptome remains relatively stable across various antiretroviral treatments and upon switching from EFV/protease inhibitor to RAL. These findings enhance our understanding of the molecular landscape on SAT in the context of HIV and ART.
Collapse
Affiliation(s)
- Anna M Hanttu
- Department of Infectious Diseases, Inflammation Center, Helsinki University Hospital and University of Helsinki
| | - Maheswary Muniandy
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine
| | - Birgitta W van der Kolk
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine
| | - Hanna Lindgren
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki
| | - Bhagwan Yadav
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki
| | - Jussi Sutinen
- Department of Infectious Diseases, Inflammation Center, Helsinki University Hospital and University of Helsinki
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine
- Healthy Weight Hub, Abdominal Center, Endocrinology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Muniandy M, Joenväärä S, van der Kolk BW, Tohmola T, Haltia H, Saari S, Hakkarainen A, Lundbom J, Kuula J, Groop PH, Kaprio J, Heinonen S, Renkonen R, Pietiläinen KH. Plasma N-Glycoproteomics in monozygotic twin pairs discordant for body mass index reveals an obesity signature related to inflammation and iron metabolism. Biol Direct 2025; 20:31. [PMID: 40108677 PMCID: PMC11921541 DOI: 10.1186/s13062-025-00609-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/20/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND N-glycosylation is a complex, post-translational modification which influences protein function and is sensitive to physiological changes. Obesity is associated with alterations in protein function; however, little is known about the glycoproteome in obesity beyond observations of association with types and structures of selected glycopeptides. Most often, due to technical challenges, glycan composition and structure information are missing. Here, we combined label-free data-independent proteomics and targeted quantitative glycoproteomics to study N-glycosylation of plasma proteins in obesity. Using a monozygotic twin study design, we controlled for genetic variation and focused only on the acquired effects of obesity. METHODS Using plasma samples of 48 monozygotic twin pairs discordant for BMI (intrapair difference > 2.5 kg/m2), we identified using mass spectrometry, differential protein and glycopeptide levels between heavier and leaner co-twins. We used a within-twin paired analysis model and considered p < 0.05 as significant. RESULTS We identified 48 protein and 33 N-glycosylation expression differences (p < 0.05) between co-twins. These differences occurred either both in the protein expression and glycoprotein (sometimes in opposing directions) or independently from each other. Haptoglobin protein was upregulated (Fold Change = 1.10, p = 0.001) in heavier co-twins along with seven upregulated glycan compositions at N-glycosylation site Asn241. The complement protein C3 was upregulated (Fold Change = 1.08, p = 0.014) along with one upregulated glycopeptide at Asn85. Additionally, many glycopeptides were upregulated despite non-significant differences in protein-backbone plasma levels. CONCLUSION Differential protein expression related to cholesterol biosynthesis and acute phase signalling as well as N-glycosylation of proteins related to iron metabolism and inflammation can be linked to acquired obesity.
Collapse
Affiliation(s)
- Maheswary Muniandy
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Sakari Joenväärä
- Transplantation Laboratory, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Birgitta W van der Kolk
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tiialotta Tohmola
- Transplantation Laboratory, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Hanna Haltia
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sina Saari
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Antti Hakkarainen
- Department of Radiology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Jesper Lundbom
- Department of Radiology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany
| | - Juho Kuula
- Department of Radiology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Faculty of Medicine Doctoral Program in Clinical Research, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Risto Renkonen
- Transplantation Laboratory, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Healthy Weight Hub, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
5
|
Florea VV, Gajjar P, Huang S, Tang J, Zhao S, Davenport M, Mi MY, Haff M, Zhang X, Miller PE, Vasan RS, Liu CT, Lewis GD, Shah RV, Long MT, Nayor M. Hepatic Steatosis and Fibrosis, Cardiorespiratory Fitness, and Metabolic Mediators in the Community. Liver Int 2025; 45:e16147. [PMID: 39673712 DOI: 10.1111/liv.16147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/25/2024] [Accepted: 10/12/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND AND AIMS Individuals with steatotic liver disease (SLD) are at high cardiovascular disease (CVD) risk, but approaches to characterise and mitigate this risk are limited. By investigating relations, and shared metabolic pathways, of hepatic steatosis/fibrosis and cardiorespiratory fitness (CRF), we sought to identify new avenues for CVD risk reduction in SLD. METHODS In Framingham Heart Study (FHS) participants (N = 2722, age 54 ± 9 years, 53% women), vibration-controlled transient elastography (VCTE) was performed between 2016-2019 to assess hepatic steatosis (continuous attenuation parameter [CAP]) and fibrosis (liver fibrosis measure [LSM]). Concurrently, participants underwent maximum effort cardiopulmonary exercise testing (CPET), and metabolomic profiling (201 circulating metabolites) was performed in a subsample (N = 1268). RESULTS Mean BMI was 28.0 ± 5.3, 27% had hepatic steatosis, 7.6% had fibrosis, and peak oxygen uptake (VO2) was 26.2 ± 6.8 mL/kg/min in men and 20.7 ± 6.0 mL/kg/min in women (95% predicted overall). In linear models adjusted for cardiometabolic risk factors, greater CAP and LSM were associated with lower peak VO2 (p ≤ 0.002 for all), and the CAP association remained significant after BMI adjustment (p < 0.0001). We observed shared metabolic architecture of CAP, LSM, and peak VO2, with metabolites mediating up to 35% (for CAP) and 74% (for LSM) of the association with peak VO2. Metabolite mediators included amino acids and derivatives implicated in cardiometabolic risk and both protective and deleterious lipid species. CONCLUSIONS Hepatic steatosis and fibrosis are associated with CRF impairment in the community, and these relations are partly mediated by pathways of altered lipid metabolism and general cardiometabolic risk.
Collapse
Affiliation(s)
- Victor V Florea
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, Massachusetts, USA
| | - Priya Gajjar
- Cardiovascular Medicine Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Shi Huang
- Vanderbilt Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jingxian Tang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Shilin Zhao
- Vanderbilt Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Megan Davenport
- Cardiovascular Medicine Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Michael Y Mi
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Madeleine Haff
- Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Xiaoyu Zhang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Patricia E Miller
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Ramachandran S Vasan
- University of Texas School of Public Health, San Antonio, Texas, USA
- Departments of Medicine and Population Health Sciences, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Gregory D Lewis
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Research Center, Cardiology Division, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michelle T Long
- Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Novo Nordisk A/S, Søborg, Denmark
| | - Matthew Nayor
- Cardiovascular Medicine Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Preventive Medicine and Epidemiology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Romero MJO, Na D. From metabolomics to energy balance physiology. ADVANCES IN GENETICS 2024; 113:102-145. [PMID: 40409795 DOI: 10.1016/bs.adgen.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Omics technologies are transforming our understanding of disease mechanisms and reshaping clinical practice. By enabling high-throughput, unbiased data collection at various molecular levels - including genes (genomics), mRNA (transcriptomics), proteins (proteomics), and metabolites (metabolomics) - omics approaches offer a comprehensive view of biological states in both health and disease. Among these, metabolomics has emerged as a pivotal tool, rapidly evolving beyond diagnostics to become a cutting-edge technique for pinpointing metabolites that regulate key physiological processes. This chapter reviews the advances in metabolomics, its integration with other omics approaches, and its applications, particularly emphasizing energy homeostasis. By incorporating metabolomic insights into physiology, we move closer to an integrative understanding of biological systems, laying the groundwork for novel therapies to combat obesity and related metabolic disorders.
Collapse
Affiliation(s)
| | - Daxiang Na
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
7
|
Meng L, Jin H, Yulug B, Altay O, Li X, Hanoglu L, Cankaya S, Coskun E, Idil E, Nogaylar R, Ozsimsek A, Shoaie S, Turkez H, Nielsen J, Zhang C, Borén J, Uhlén M, Mardinoglu A. Multi-omics analysis reveals the key factors involved in the severity of the Alzheimer's disease. Alzheimers Res Ther 2024; 16:213. [PMID: 39358810 PMCID: PMC11448018 DOI: 10.1186/s13195-024-01578-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder with a global impact, yet its pathogenesis remains poorly understood. While age, metabolic abnormalities, and accumulation of neurotoxic substances are potential risk factors for AD, their effects are confounded by other factors. To address this challenge, we first utilized multi-omics data from 87 well phenotyped AD patients and generated plasma proteomics and metabolomics data, as well as gut and saliva metagenomics data to investigate the molecular-level alterations accounting the host-microbiome interactions. Second, we analyzed individual omics data and identified the key parameters involved in the severity of the dementia in AD patients. Next, we employed Artificial Intelligence (AI) based models to predict AD severity based on the significantly altered features identified in each omics analysis. Based on our integrative analysis, we found the clinical relevance of plasma proteins, including SKAP1 and NEFL, plasma metabolites including homovanillate and glutamate, and Paraprevotella clara in gut microbiome in predicting the AD severity. Finally, we validated the predictive power of our AI based models by generating additional multi-omics data from the same group of AD patients by following up for 3 months. Hence, we observed that these results may have important implications for the development of potential diagnostic and therapeutic approaches for AD patients.
Collapse
Affiliation(s)
- Lingqi Meng
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Han Jin
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Burak Yulug
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Ozlem Altay
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Xiangyu Li
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Lutfu Hanoglu
- Department of Neurology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Seyda Cankaya
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Ebru Coskun
- Department of Neurology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ezgi Idil
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Rahim Nogaylar
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Ahmet Ozsimsek
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Saeed Shoaie
- Centre for Host-Microbiome Interaction's, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden.
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden.
- Centre for Host-Microbiome Interaction's, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK.
| |
Collapse
|
8
|
Fang L, Liu C, Jiang ZZ, Wang M, Geng K, Xu Y, Zhu Y, Fu Y, Xue J, Shan W, Zhang Q, Chen J, Chen J, Zhao M, Guo Y, Siu KWM, Chen YE, Xu Y, Liu D, Zheng L. Annexin A1 binds PDZ and LIM domain 7 to inhibit adipogenesis and prevent obesity. Signal Transduct Target Ther 2024; 9:218. [PMID: 39174522 PMCID: PMC11341699 DOI: 10.1038/s41392-024-01930-0] [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: 01/10/2024] [Revised: 06/29/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024] Open
Abstract
Obesity is a global issue that warrants the identification of more effective therapeutic targets and a better understanding of the pivotal molecular pathogenesis. Annexin A1 (ANXA1) is known to inhibit phospholipase A2, exhibiting anti-inflammatory activity. However, the specific effects of ANXA1 in obesity and the underlying mechanisms of action remain unclear. Our study reveals that ANXA1 levels are elevated in the adipose tissue of individuals with obesity. Whole-body or adipocyte-specific ANXA1 deletion aggravates obesity and metabolic disorders. ANXA1 levels are higher in stromal vascular fractions (SVFs) than in mature adipocytes. Further investigation into the role of ANXA1 in SVFs reveals that ANXA1 overexpression induces lower numbers of mature adipocytes, while ANXA1-knockout SVFs exhibit the opposite effect. This suggests that ANXA1 plays an important role in adipogenesis. Mechanistically, ANXA1 competes with MYC binding protein 2 (MYCBP2) for interaction with PDZ and LIM domain 7 (PDLIM7). This exposes the MYCBP2-binding site, allowing it to bind more readily to the SMAD family member 4 (SMAD4) and promoting its ubiquitination and degradation. SMAD4 degradation downregulates peroxisome proliferator-activated receptor gamma (PPARγ) transcription and reduces adipogenesis. Treatment with Ac2-26, an active peptide derived from ANXA1, inhibits both adipogenesis and obesity through the mechanism. In conclusion, the molecular mechanism of ANXA1 inhibiting adipogenesis was first uncovered in our study, which is a potential target for obesity prevention and treatment.
Collapse
Affiliation(s)
- Lu Fang
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 100191, Beijing, China
| | - Changjie Liu
- Department of Blood Transfusion, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, Guangdong, China
| | - Zong-Zhe Jiang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China
| | - Mengxiao Wang
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 100191, Beijing, China
| | - Kang Geng
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China
- Department of plastic and burns surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, PR China
| | - Yangkai Xu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 100191, Beijing, China
| | - Yujie Zhu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 100191, Beijing, China
| | - Yiwen Fu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 100191, Beijing, China
| | - Jing Xue
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Capital Medical University, 6 Tiantan Xili, Chongwen District, 100050, Beijing, China
| | - Wenxin Shan
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 100191, Beijing, China
| | - Qi Zhang
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 100191, Beijing, China
| | - Jie Chen
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 100191, Beijing, China
| | - Jiahong Chen
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 100191, Beijing, China
| | - Mingming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, 100191, Beijing, China
| | - Yuxuan Guo
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 100191, Beijing, China
| | - K W Michael Siu
- Center for Mass Spectrometry Research and Clinical Application, Shandong Public Health Clinical Center Affiliated to Shandong University, Lishan Campus, 46 Lishan Road, Jinan, Shandong, China
- Department of Chemistry and Biochemistry, University of Windsor, 401 Sunset Avenue, Windsor, ON, N9B 3P4, Canada
| | - Y Eugene Chen
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, PR China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China.
| | - Donghui Liu
- Department of Geriatrics, National Key Clinical Specialty, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, China.
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 100191, Beijing, China.
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Capital Medical University, 6 Tiantan Xili, Chongwen District, 100050, Beijing, China.
| |
Collapse
|
9
|
Bogaards FA, Gehrmann T, Beekman M, Lakenberg N, Suchiman HED, de Groot CPGM, Reinders MJT, Slagboom PE. Secondary integrated analysis of multi-tissue transcriptomic responses to a combined lifestyle intervention in older adults from the GOTO nonrandomized trial. Nat Commun 2024; 15:7013. [PMID: 39147741 PMCID: PMC11327278 DOI: 10.1038/s41467-024-50693-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 07/18/2024] [Indexed: 08/17/2024] Open
Abstract
Molecular effects of lifestyle interventions are typically studied in a single tissue. Here, we perform a secondary analysis on the sex-specific effects of the Growing Old TOgether trial (GOTO, trial registration number GOT NL3301 ( https://onderzoekmetmensen.nl/nl/trial/27183 ), NL-OMON27183 , primary outcomes have been previously reported in ref. 1), a moderate 13-week combined lifestyle intervention on the transcriptomes of postprandial blood, subcutaneous adipose tissue (SAT) and muscle tissue in healthy older adults, the overlap in effect between tissues and their relation to whole-body parameters of metabolic health. The GOTO intervention has virtually no effect on the postprandial blood transcriptome, while the SAT and muscle transcriptomes respond significantly. In SAT, pathways involved in HDL remodeling, O2/CO2 exchange and signaling are overrepresented, while in muscle, collagen and extracellular matrix pathways are significantly overexpressed. Additionally, we find that the effects of the SAT transcriptome closest associates with gains in metabolic health. Lastly, in males, we identify a shared variation between the transcriptomes of the three tissues. We conclude that the GOTO intervention has a significant effect on metabolic and muscle fibre pathways in the SAT and muscle transcriptome, respectively. Aligning the response in the three tissues revealed a blood transcriptome component which may act as an integrated health marker for metabolic intervention effects across tissues.
Collapse
Affiliation(s)
- F A Bogaards
- Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands.
- Leiden Computational Biology Center, Leiden, The Netherlands.
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands.
| | - T Gehrmann
- Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Computational Biology Center, Leiden, The Netherlands
- Department of Bioscience Engineering, Lab of Applied Microbiology and Biotechnology, University of Antwerp, Antwerp, Belgium
| | - M Beekman
- Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - N Lakenberg
- Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - H E D Suchiman
- Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - C P G M de Groot
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - M J T Reinders
- Leiden Computational Biology Center, Leiden, The Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
| | - P E Slagboom
- Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
- Max Planck Institute for Biology of Aging, Cologne, Germany
| |
Collapse
|
10
|
Zhang J, Liang D, Xu L, Liu Y, Jiang S, Han X, Wu H, Jiang Y. Associations between novel anthropometric indices and the prevalence of gallstones among 6,848 adults: a cross-sectional study. Front Nutr 2024; 11:1428488. [PMID: 39104753 PMCID: PMC11298442 DOI: 10.3389/fnut.2024.1428488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024] Open
Abstract
Background Traditional anthropometric measures, including body mass index (BMI), are insufficient for evaluating gallstone risk. This study investigated the association between novel anthropometric indices and gallstone risk among 6,848 participants from the National Health and Nutrition Examination Survey in the United States. Methods Measures calculated included weight (WT), BMI, waist circumference (WC), waist-to-height ratio (WtHR), conicity index (CI), A Body Shape Index (ABSI), Body Roundness Index (BRI), Abdominal Volume Index (AVI), and Weight-adjusted Waist Index (WWI). Logistic regression and smooth curve fitting assessed the relationships between these indices and gallstones, complemented by receiver operating characteristic (ROC) curve analysis to evaluate their discriminative power. Results The results indicated significant differences between study groups, with a positive and independent correlation identified between gallstones and all measures except ABSI. Specifically, per 1 SD increase in WC, WT, BMI, WtHR, and AVI was associated with a 57%, 59%, 52%, 53%, and 53% increased risk of gallstones, respectively. Dose-response analysis confirmed a positive correlation between these indices and gallstone risk. ROC analysis highlighted WtHR and BRI as having superior discriminative abilities (AUC = 0.6703). Further, among participants with a BMI < 30 kg/m2, elevated levels of WT, WtHR, CI, BRI, and WWI significantly increased the risk of gallstones (P < 0.001). Likewise, elevated BMI heightened the risk at low levels of WT, WC, WtHR, BRI, AVI, and CI (P < 0.001). Conclusion This study supports the positive association between various anthropometric indicators and gallstones, recommending that newer anthropometric indices be considered more extensively to enhance gallstone prevention and treatment strategies.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan Provincial Medicine Key Laboratory of Colorectal Cancer Diagnosis and Treatment, Zhengzhou, China
- Zhengzhou Key Laboratory of Colorectal Cancer Diagnosis, Treatment and Research, Zhengzhou, China
| | - Depeng Liang
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan Provincial Medicine Key Laboratory of Colorectal Cancer Diagnosis and Treatment, Zhengzhou, China
- Zhengzhou Key Laboratory of Colorectal Cancer Diagnosis, Treatment and Research, Zhengzhou, China
| | - Lidong Xu
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan Provincial Medicine Key Laboratory of Colorectal Cancer Diagnosis and Treatment, Zhengzhou, China
- Zhengzhou Key Laboratory of Colorectal Cancer Diagnosis, Treatment and Research, Zhengzhou, China
| | - Yanhong Liu
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan Provincial Medicine Key Laboratory of Colorectal Cancer Diagnosis and Treatment, Zhengzhou, China
- Zhengzhou Key Laboratory of Colorectal Cancer Diagnosis, Treatment and Research, Zhengzhou, China
| | - Shan Jiang
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan Provincial Medicine Key Laboratory of Colorectal Cancer Diagnosis and Treatment, Zhengzhou, China
- Zhengzhou Key Laboratory of Colorectal Cancer Diagnosis, Treatment and Research, Zhengzhou, China
| | - Xiaomeng Han
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan Provincial Medicine Key Laboratory of Colorectal Cancer Diagnosis and Treatment, Zhengzhou, China
- Zhengzhou Key Laboratory of Colorectal Cancer Diagnosis, Treatment and Research, Zhengzhou, China
| | - Huili Wu
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan Provincial Medicine Key Laboratory of Colorectal Cancer Diagnosis and Treatment, Zhengzhou, China
- Zhengzhou Key Laboratory of Colorectal Cancer Diagnosis, Treatment and Research, Zhengzhou, China
| | - Yuanyuan Jiang
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan Provincial Medicine Key Laboratory of Colorectal Cancer Diagnosis and Treatment, Zhengzhou, China
- Zhengzhou Key Laboratory of Colorectal Cancer Diagnosis, Treatment and Research, Zhengzhou, China
| |
Collapse
|
11
|
Tan J, Virtue S, Norris DM, Conway OJ, Yang M, Bidault G, Gribben C, Lugtu F, Kamzolas I, Krycer JR, Mills RJ, Liang L, Pereira C, Dale M, Shun-Shion AS, Baird HJ, Horscroft JA, Sowton AP, Ma M, Carobbio S, Petsalaki E, Murray AJ, Gershlick DC, Nathan JA, Hudson JE, Vallier L, Fisher-Wellman KH, Frezza C, Vidal-Puig A, Fazakerley DJ. Limited oxygen in standard cell culture alters metabolism and function of differentiated cells. EMBO J 2024; 43:2127-2165. [PMID: 38580776 PMCID: PMC11148168 DOI: 10.1038/s44318-024-00084-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/20/2024] [Accepted: 03/03/2024] [Indexed: 04/07/2024] Open
Abstract
The in vitro oxygen microenvironment profoundly affects the capacity of cell cultures to model physiological and pathophysiological states. Cell culture is often considered to be hyperoxic, but pericellular oxygen levels, which are affected by oxygen diffusivity and consumption, are rarely reported. Here, we provide evidence that several cell types in culture actually experience local hypoxia, with important implications for cell metabolism and function. We focused initially on adipocytes, as adipose tissue hypoxia is frequently observed in obesity and precedes diminished adipocyte function. Under standard conditions, cultured adipocytes are highly glycolytic and exhibit a transcriptional profile indicative of physiological hypoxia. Increasing pericellular oxygen diverted glucose flux toward mitochondria, lowered HIF1α activity, and resulted in widespread transcriptional rewiring. Functionally, adipocytes increased adipokine secretion and sensitivity to insulin and lipolytic stimuli, recapitulating a healthier adipocyte model. The functional benefits of increasing pericellular oxygen were also observed in macrophages, hPSC-derived hepatocytes and cardiac organoids. Our findings demonstrate that oxygen is limiting in many terminally-differentiated cell types, and that considering pericellular oxygen improves the quality, reproducibility and translatability of culture models.
Collapse
Affiliation(s)
- Joycelyn Tan
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Sam Virtue
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Dougall M Norris
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Olivia J Conway
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Ming Yang
- MRC Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
- CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Cologne, 50931, Germany
| | - Guillaume Bidault
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Christopher Gribben
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Fatima Lugtu
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Ioannis Kamzolas
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - James R Krycer
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, 4000, Australia
| | - Richard J Mills
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, 4000, Australia
| | - Lu Liang
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Conceição Pereira
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Martin Dale
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Amber S Shun-Shion
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Harry Jm Baird
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - James A Horscroft
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EL, UK
| | - Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EL, UK
| | - Marcella Ma
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Stefania Carobbio
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
- Centro de Investigacion Principe Felipe, Valencia, 46012, Spain
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EL, UK
| | - David C Gershlick
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - James A Nathan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - James E Hudson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, 4000, Australia
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ludovic Vallier
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Kelsey H Fisher-Wellman
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Christian Frezza
- MRC Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
- CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Cologne, 50931, Germany
| | - Antonio Vidal-Puig
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Centro de Investigacion Principe Felipe, Valencia, 46012, Spain.
| | - Daniel J Fazakerley
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
12
|
Antonatos C, Georgakilas GK, Evangelou E, Vasilopoulos Y. Transcriptomic meta-analysis characterizes molecular commonalities between psoriasis and obesity. Genes Immun 2024; 25:179-187. [PMID: 38580831 DOI: 10.1038/s41435-024-00271-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024]
Abstract
Despite the abundance of epidemiological evidence for the high comorbid rate between psoriasis and obesity, systematic approaches to common inflammatory mechanisms have not been adequately explored. We performed a meta-analysis of publicly available RNA-sequencing datasets to unveil putative mechanisms that are postulated to exacerbate both diseases, utilizing both late-stage, disease-specific meta-analyses and consensus gene co-expression network (cWGCNA). Single-gene meta-analyses reported several common inflammatory mechanisms fostered by the perturbed expression profile of inflammatory cells. Assessment of gene overlaps between both diseases revealed significant overlaps between up- (n = 170, P value = 6.07 × 10-65) and down-regulated (n = 49, P value = 7.1 × 10-7) genes, associated with increased T cell response and activated transcription factors. Our cWGCNA approach disentangled 48 consensus modules, associated with either the differentiation of leukocytes or metabolic pathways with similar correlation signals in both diseases. Notably, all our analyses confirmed the association of the perturbed T helper (Th)17 differentiation pathway in both diseases. Our novel findings through whole transcriptomic analyses characterize the inflammatory commonalities between psoriasis and obesity implying the assessment of several expression profiles that could serve as putative comorbid disease progression biomarkers and therapeutic interventions.
Collapse
Affiliation(s)
- Charalabos Antonatos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504, Patras, Greece
| | - Georgios K Georgakilas
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504, Patras, Greece
- Information Management Systems Institute (IMSI), ATHENA Research Center, 15125, Athens, Greece
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, 45110, Ioannina, Greece
- Department of Epidemiology & Biostatistics, MRC Centre for Environment and Health, Imperial College London, London, W2 1PG, UK
| | - Yiannis Vasilopoulos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504, Patras, Greece.
| |
Collapse
|
13
|
Das SS, Das SK. Common and ethnic-specific derangements in skeletal muscle transcriptome associated with obesity. Int J Obes (Lond) 2024; 48:330-338. [PMID: 37993634 DOI: 10.1038/s41366-023-01417-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/25/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Obesity is a common disease with a higher prevalence among African Americans. Obesity alters cellular function in many tissues, including skeletal muscle, and is a risk factor for many life-threatening diseases, including cardiovascular disease and diabetes. The similarities and differences in molecular mechanisms that may explain ethnic disparities in obesity between African and European ancestry individuals have not been studied. METHODS In this study, data from transcriptome-wide analyses on skeletal muscle tissues from well-powered human cohorts were used to compare genes and biological pathways affected by obesity in European and African ancestry populations. Data on obesity-induced differentially expressed transcripts and GWAS-identified SNPs were integrated to prioritize target genes for obesity-associated genetic variants. RESULTS Linear regression analysis in the FUSION (European, N = 301) and AAGMEx (African American, N = 256) cohorts identified a total of 2569 body mass index (BMI)-associated transcripts (q < 0.05), of which 970 genes (at p < 0.05) are associated in both cohorts, and the majority showed the same direction of effect on BMI. Biological pathway analyses, including over-representation and gene-set enrichment analyses, identified enrichment of protein synthesis pathways (e.g., ribosomal function) and the ceramide signaling pathway in both cohorts among BMI-associated down- and up-regulated transcripts, respectively. A comparison using the IPA-tool suggested the activation of inflammation pathways only in Europeans with obesity. Interestingly, these analyses suggested repression of the mitochondrial oxidative phosphorylation pathway in Europeans but showed its activation in African Americans. Integration of SNP-to-Gene analyses-predicted target genes for obesity-associated genetic variants (GWAS-identified SNPs) and BMI-associated transcripts suggested that these SNPs might cause obesity by altering the expression of 316 critical target genes (e.g., GRB14) in the muscle. CONCLUSIONS This study provides a replication of obesity-associated transcripts and biological pathways in skeletal muscle across ethnicities, but also identifies obesity-associated processes unique in either African or European ancestry populations.
Collapse
Affiliation(s)
- Sreejon S Das
- The School of Biotechnology at Atkins, Atkins Academic and Technology High, Winston-Salem, NC, 27101, USA
| | - Swapan K Das
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
14
|
Ouni M, Kovac L, Gancheva S, Jähnert M, Zuljan E, Gottmann P, Kahl S, de Angelis MH, Roden M, Schürmann A. Novel markers and networks related to restored skeletal muscle transcriptome after bariatric surgery. Obesity (Silver Spring) 2024; 32:363-375. [PMID: 38086776 DOI: 10.1002/oby.23954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 01/26/2024]
Abstract
OBJECTIVE The aim of this study was to discover novel markers underlying the improvement of skeletal muscle metabolism after bariatric surgery. METHODS Skeletal muscle transcriptome data of lean people and people with obesity, before and 1 year after bariatric surgery, were subjected to weighted gene co-expression network analysis (WGCNA) and least absolute shrinkage and selection operator (LASSO) regression. Results of LASSO were confirmed in a replication cohort. RESULTS The expression levels of 440 genes differing between individuals with and without obesity were no longer different 1 year after surgery, indicating restoration. WGCNA clustered 116 genes with normalized expression in one major module, particularly correlating to weight loss and decreased plasma free fatty acids (FFA), 44 of which showed an obesity-related phenotype upon deletion in mice. Among the genes of the major module, 105 represented prominent markers for reduced FFA concentration, including 55 marker genes for decreased BMI in both the discovery and replication cohorts. CONCLUSIONS Previously unknown gene networks and marker genes underlined the important role of FFA in restoring muscle gene expression after bariatric surgery and further suggest novel therapeutic targets for obesity.
Collapse
Affiliation(s)
- Meriem Ouni
- German Institute of Human Nutrition, Department of Experimental Diabetology, Potsdam, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
| | - Leona Kovac
- German Institute of Human Nutrition, Department of Experimental Diabetology, Potsdam, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
| | - Sofiya Gancheva
- German Center for Diabetes Research (DZD), Munich, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany
| | - Markus Jähnert
- German Institute of Human Nutrition, Department of Experimental Diabetology, Potsdam, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
| | - Erika Zuljan
- German Institute of Human Nutrition, Department of Experimental Diabetology, Potsdam, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
| | - Pascal Gottmann
- German Institute of Human Nutrition, Department of Experimental Diabetology, Potsdam, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
| | - Sabine Kahl
- German Center for Diabetes Research (DZD), Munich, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Martin Hrabĕ de Angelis
- German Center for Diabetes Research (DZD), Munich, Germany
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- School of Life Sciences, Technical University Munich, Freising, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD), Munich, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany
| | - Annette Schürmann
- German Institute of Human Nutrition, Department of Experimental Diabetology, Potsdam, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
- Institute of Nutritional Sciences, University of Potsdam, Nuthetal, Germany
| |
Collapse
|
15
|
Larsen JK, Kruse R, Sahebekhtiari N, Moreno-Justicia R, Gomez Jorba G, Petersen MH, de Almeida ME, Ørtenblad N, Deshmukh AS, Højlund K. High-throughput proteomics uncovers exercise training and type 2 diabetes-induced changes in human white adipose tissue. SCIENCE ADVANCES 2023; 9:eadi7548. [PMID: 38019916 PMCID: PMC10686561 DOI: 10.1126/sciadv.adi7548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023]
Abstract
White adipose tissue (WAT) is important for metabolic homeostasis. We established the differential proteomic signatures of WAT in glucose-tolerant lean and obese individuals and patients with type 2 diabetes (T2D) and the response to 8 weeks of high-intensity interval training (HIIT). Using a high-throughput and reproducible mass spectrometry-based proteomics pipeline, we identified 3773 proteins and found that most regulated proteins displayed progression in markers of dysfunctional WAT from lean to obese to T2D individuals and were highly associated with clinical measures such as insulin sensitivity and HbA1c. We propose that these distinct markers could serve as potential clinical biomarkers. HIIT induced only minor changes in the WAT proteome. This included an increase in WAT ferritin levels independent of obesity and T2D, and WAT ferritin levels were strongly correlated with individual insulin sensitivity. Together, we report a proteomic signature of WAT related to obesity and T2D and highlight an unrecognized role of human WAT iron metabolism in exercise training adaptations.
Collapse
Affiliation(s)
- Jeppe Kjærgaard Larsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Kruse
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Navid Sahebekhtiari
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Roger Moreno-Justicia
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Gerard Gomez Jorba
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Maria H. Petersen
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark
| | - Martin E. de Almeida
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Niels Ørtenblad
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Atul S. Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
16
|
Sabaratnam R, Hansen DR, Svenningsen P. White adipose tissue mitochondrial bioenergetics in metabolic diseases. Rev Endocr Metab Disord 2023; 24:1121-1133. [PMID: 37558853 DOI: 10.1007/s11154-023-09827-z] [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] [Accepted: 07/19/2023] [Indexed: 08/11/2023]
Abstract
White adipose tissue (WAT) is an important endocrine organ that regulates systemic energy metabolism. In metabolically unhealthy obesity, adipocytes become dysfunctional through hypertrophic mechanisms associated with a reduced endocrine function, reduced mitochondrial function, but increased inflammation, fibrosis, and extracellular remodelling. A pathologic WAT remodelling promotes systemic lipotoxicity characterized by fat accumulation in tissues such as muscle and liver, leading to systemic insulin resistance and type 2 diabetes. Several lines of evidence from human and animal studies suggest a link between unhealthy obesity and adipocyte mitochondrial dysfunction, and interventions that improve mitochondrial function may reduce the risk of obesity-associated diseases. This review discusses the importance of mitochondrial function and metabolism in human adipocyte biology and intercellular communication mechanisms within WAT. Moreover, a selected interventional approach for better adipocyte mitochondrial metabolism in humans is reviewed. A greater understanding of mitochondrial bioenergetics in WAT might provide novel therapeutic opportunities to prevent or restore dysfunctional adipose tissue in obesity-associated diseases.
Collapse
Affiliation(s)
- Rugivan Sabaratnam
- Department of Clinical Research, University of Southern Denmark, Odense C, DK-5000, Denmark.
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, DK-5000, Denmark.
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark.
| | - Didde Riisager Hansen
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, DK-5000, Denmark
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark
| | - Per Svenningsen
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark.
| |
Collapse
|
17
|
Zoh RS, Yu X, Dawid P, Smith GD, French SJ, Allison DB. Causal models and causal modelling in obesity: foundations, methods and evidence. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220227. [PMID: 37661742 PMCID: PMC10475873 DOI: 10.1098/rstb.2022.0227] [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: 03/17/2023] [Accepted: 08/06/2023] [Indexed: 09/05/2023] Open
Abstract
Discussing causes in science, if we are to do so in a way that is sensible, begins at the root. All too often, we jump to discussing specific postulated causes but do not first consider what we mean by, for example, causes of obesity or how we discern whether something is a cause. In this paper, we address what we mean by a cause, discuss what might and might not constitute a reasonable causal model in the abstract, speculate about what the causal structure of obesity might be like overall and the types of things we should be looking for, and finally, delve into methods for evaluating postulated causes and estimating causal effects. We offer the view that different meanings of the concept of causal factors in obesity research are regularly being conflated, leading to confusion, unclear thinking and sometimes nonsense. We emphasize the idea of different kinds of studies for evaluating various aspects of causal effects and discuss experimental methods, assumptions and evaluations. We use analogies from other areas of research to express the plausibility that only inelegant solutions will be truly informative. Finally, we offer comments on some specific postulated causal factors. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part II)'.
Collapse
Affiliation(s)
- Roger S. Zoh
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Bloomington, IN, 47405-7000, USA
| | - Xiaoxin Yu
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Bloomington, IN, 47405-7000, USA
| | | | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Bristol, UK
| | - Stephen J. French
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Bloomington, IN, 47405-7000, USA
| | - David B. Allison
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Bloomington, IN, 47405-7000, USA
| |
Collapse
|
18
|
Das SS, Kar A, Rajkumar S, Lee SHT, Alvarez M, Pietiläinen KH, Pajukanta P. Cross-Tissue Single-Nucleus RNA Sequencing Discovers Tissue-Resident Adipocytes Involved in Propanoate Metabolism in the Human Heart. Arterioscler Thromb Vasc Biol 2023; 43:1788-1804. [PMID: 37409528 PMCID: PMC10538422 DOI: 10.1161/atvbaha.123.319358] [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: 04/04/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Adipocytes are crucial regulators of cardiovascular health. However, not much is known about gene expression profiles of adipocytes residing in nonfat cardiovascular tissues, their genetic regulation, and contribution to coronary artery disease. Here, we investigated whether and how the gene expression profiles of adipocytes in the subcutaneous adipose tissue differ from adipocytes residing in the heart. METHODS We used single-nucleus RNA-sequencing data sets of subcutaneous adipose tissue and heart and performed in-depth analysis of tissue-resident adipocytes and their cell-cell interactions. RESULTS We first discovered tissue-specific features of tissue-resident adipocytes, identified functional pathways involved in their tissue specificity, and found genes with cell type-specific expression enrichment in tissue-resident adipocytes. By following up these results, we discovered the propanoate metabolism pathway as a novel distinct characteristic of the heart-resident adipocytes and found a significant enrichment of coronary artery disease genome-wide association study risk variants among the right atrium-specific adipocyte marker genes. Our cell-cell communication analysis identified 22 specific heart adipocyte-associated ligand-receptor pairs and signaling pathways, including THBS (thrombospondin) and EPHA (ephrin type-A), further supporting the distinct tissue-resident role of heart adipocytes. Our results also suggest chamber-level coordination of heart adipocyte expression profiles as we observed a consistently larger number of adipocyte-associated ligand-receptor interactions and functional pathways in the atriums than ventricles. CONCLUSIONS Overall, we introduce a new function and genetic link to coronary artery disease for the previously unexplored heart-resident adipocytes.
Collapse
Affiliation(s)
- Sankha Subhra Das
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Asha Kar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Sandhya Rajkumar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Seung Hyuk T. Lee
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HealthyWeightHub, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, USA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, USA
| |
Collapse
|
19
|
Jersin RÅ, Sri Priyanka Tallapragada D, Skartveit L, Bjune MS, Muniandy M, Lee-Ødegård S, Heinonen S, Alvarez M, Birkeland KI, André Drevon C, Pajukanta P, McCann A, Pietiläinen KH, Claussnitzer M, Mellgren G, Dankel SN. Impaired Adipocyte SLC7A10 Promotes Lipid Storage in Association With Insulin Resistance and Altered BCAA Metabolism. J Clin Endocrinol Metab 2023; 108:2217-2229. [PMID: 36916878 PMCID: PMC10438883 DOI: 10.1210/clinem/dgad148] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023]
Abstract
CONTEXT The neutral amino acid transporter SLC7A10/ASC-1 is an adipocyte-expressed gene with reduced expression in insulin resistance and obesity. Inhibition of SLC7A10 in adipocytes was shown to increase lipid accumulation despite decreasing insulin-stimulated uptake of glucose, a key substrate for de novo lipogenesis. These data imply that alternative lipogenic substrates to glucose fuel continued lipid accumulation during insulin resistance in obesity. OBJECTIVE We examined whether increased lipid accumulation during insulin resistance in adipocytes may involve alter flux of lipogenic amino acids dependent on SLC7A10 expression and activity, and whether this is reflected by extracellular and circulating concentrations of marker metabolites. METHODS In adipocyte cultures with impaired SLC7A10, we performed RNA sequencing and relevant functional assays. By targeted metabolite analyses (GC-MS/MS), flux of all amino acids and selected metabolites were measured in human and mouse adipose cultures. Additionally, SLC7A10 mRNA levels in human subcutaneous adipose tissue (SAT) were correlated to candidate metabolites and adiposity phenotypes in 2 independent cohorts. RESULTS SLC7A10 impairment altered expression of genes related to metabolic processes, including branched-chain amino acid (BCAA) catabolism, lipogenesis, and glyceroneogenesis. In 3T3-L1 adipocytes, SLC7A10 inhibition increased fatty acid uptake and cellular content of glycerol and cholesterol. SLC7A10 impairment in SAT cultures altered uptake of aspartate and glutamate, and increased net uptake of BCAAs, while increasing the net release of the valine catabolite 3- hydroxyisobutyrate (3-HIB). In human cohorts, SLC7A10 mRNA correlated inversely with total fat mass, circulating triacylglycerols, BCAAs, and 3-HIB. CONCLUSION Reduced SLC7A10 activity strongly affects flux of BCAAs in adipocytes, which may fuel continued lipogenesis during insulin resistance, and be reflected in increased circulating levels of the valine-derived catabolite 3-HIB.
Collapse
Affiliation(s)
- Regine Å Jersin
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Divya Sri Priyanka Tallapragada
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Linn Skartveit
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Mona S Bjune
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Maheswary Muniandy
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Sindre Lee-Ødegård
- Department of Transplantation Medicine, The University of Oslo, Institute of Clinical Medicine, and Oslo University Hospital, N-0372 Oslo, Norway
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Kåre Inge Birkeland
- Department of Transplantation Medicine, The University of Oslo, Institute of Clinical Medicine, and Oslo University Hospital, N-0372 Oslo, Norway
| | - Christian André Drevon
- Department of Nutrition, The University of Oslo, Institute of Basic Medical Sciences, N-0372 Oslo, Norway
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA 90095, USA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Adrian McCann
- Bevital A/S, Laboratoriebygget, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
- Obesity Center, Endocrinology, Abdominal Center, Helsinki University Hospital and University of Helsinki, FIN-00014 Helsinki, Finland
| | - Melina Claussnitzer
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Simon N Dankel
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| |
Collapse
|
20
|
Burton MA, Antoun E, Garratt ES, Westbury L, Baczynska A, Dennison EM, Harvey NC, Cooper C, Patel HP, Godfrey KM, Lillycrop KA, EpiGen Global Research Consortium. Adiposity is associated with widespread transcriptional changes and downregulation of longevity pathways in aged skeletal muscle. J Cachexia Sarcopenia Muscle 2023; 14:1762-1774. [PMID: 37199333 PMCID: PMC10401538 DOI: 10.1002/jcsm.13255] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 03/03/2023] [Accepted: 04/15/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND Amongst healthy older people, a number of correlates of impaired skeletal muscle mass and function have been defined. Although the prevalence of obesity is increasing markedly in this age group, information is sparse about the particular impacts of obesity on ageing skeletal muscle or the molecular mechanisms that underlie this and associated disease risk. METHODS Here, we examined genome-wide transcriptional changes using RNA sequencing in muscle biopsies from 40 older community-dwelling men from the Hertfordshire Sarcopenia Study with regard to obesity (body mass index [BMI] >30 kg/m2 , n = 7), overweight (BMI 25-30, n = 19), normal weight (BMI < 25, n = 14), and per cent and total fat mass. In addition, we used EPIC DNA methylation array data to investigate correlations between DNA methylation and gene expression in aged skeletal muscle tissue and investigated the relationship between genes within altered regulatory pathways and muscle histological parameters. RESULTS Individuals with obesity demonstrated a prominent modified transcriptional signature in muscle tissue, with a total of 542 differentially expressed genes associated with obesity (false discovery rate ≤0.05), of which 425 genes were upregulated when compared with normal weight. Upregulated genes were enriched in immune response (P = 3.18 × 10-41 ) and inflammation (leucocyte activation, P = 1.47 × 10-41 ; tumour necrosis factor, P = 2.75 × 10-15 ) signalling pathways and downregulated genes enriched in longevity (P = 1.5 × 10-3 ) and AMP-activated protein kinase (AMPK) (P = 4.5 × 10-3 ) signalling pathways. Furthermore, differentially expressed genes in both longevity and AMPK signalling pathways were associated with a change in DNA methylation, with a total of 256 and 360 significant cytosine-phosphate-guanine-gene correlations identified, respectively. Similar changes in the muscle transcriptome were observed with respect to per cent fat mass and total fat mass. Obesity was further associated with a significant increase in type II fast-fibre area (P = 0.026), of which key regulatory genes within both longevity and AMPK pathways were significantly associated. CONCLUSIONS We provide for the first time a global transcriptomic profile of skeletal muscle in older people with and without obesity, demonstrating modulation of key genes and pathways implicated in the regulation of muscle function, changes in DNA methylation associated with such pathways and associations between genes within the modified pathways implicated in muscle regulation and changes in muscle fibre type.
Collapse
Affiliation(s)
- Mark A. Burton
- Human Development and Health Academic Unit, Faculty of MedicineUniversity of SouthamptonSouthamptonUK
| | - Elie Antoun
- Human Development and Health Academic Unit, Faculty of MedicineUniversity of SouthamptonSouthamptonUK
- Biological SciencesUniversity of SouthamptonSouthamptonUK
| | - Emma S. Garratt
- Human Development and Health Academic Unit, Faculty of MedicineUniversity of SouthamptonSouthamptonUK
- National Institute for Health Research Southampton Biomedical Research CentreUniversity of Southampton and University Hospital Southampton NHS Foundation TrustSouthamptonUK
| | - Leo Westbury
- MRC Lifecourse Epidemiology CentreUniversity of SouthamptonSouthamptonUK
| | - Alica Baczynska
- Academic Geriatric Medicine, Faculty of MedicineUniversity of SouthamptonSouthamptonUK
| | - Elaine M. Dennison
- MRC Lifecourse Epidemiology CentreUniversity of SouthamptonSouthamptonUK
- Victoria University of WellingtonWellingtonNew Zealand
| | - Nicholas C. Harvey
- National Institute for Health Research Southampton Biomedical Research CentreUniversity of Southampton and University Hospital Southampton NHS Foundation TrustSouthamptonUK
- MRC Lifecourse Epidemiology CentreUniversity of SouthamptonSouthamptonUK
| | - Cyrus Cooper
- National Institute for Health Research Southampton Biomedical Research CentreUniversity of Southampton and University Hospital Southampton NHS Foundation TrustSouthamptonUK
- MRC Lifecourse Epidemiology CentreUniversity of SouthamptonSouthamptonUK
- NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| | - Harnish P. Patel
- National Institute for Health Research Southampton Biomedical Research CentreUniversity of Southampton and University Hospital Southampton NHS Foundation TrustSouthamptonUK
- MRC Lifecourse Epidemiology CentreUniversity of SouthamptonSouthamptonUK
- Academic Geriatric Medicine, Faculty of MedicineUniversity of SouthamptonSouthamptonUK
| | - Keith M. Godfrey
- Human Development and Health Academic Unit, Faculty of MedicineUniversity of SouthamptonSouthamptonUK
- MRC Lifecourse Epidemiology CentreUniversity of SouthamptonSouthamptonUK
- Academic Geriatric Medicine, Faculty of MedicineUniversity of SouthamptonSouthamptonUK
| | - Karen A. Lillycrop
- Human Development and Health Academic Unit, Faculty of MedicineUniversity of SouthamptonSouthamptonUK
- Biological SciencesUniversity of SouthamptonSouthamptonUK
- National Institute for Health Research Southampton Biomedical Research CentreUniversity of Southampton and University Hospital Southampton NHS Foundation TrustSouthamptonUK
| | | |
Collapse
|
21
|
Proteomic Analysis of Skeletal Muscle and White Adipose Tissue after Aerobic Exercise Training in High Fat Diet Induced Obese Mice. Int J Mol Sci 2023; 24:ijms24065743. [PMID: 36982812 PMCID: PMC10052314 DOI: 10.3390/ijms24065743] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/08/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Obesity is associated with excessive fat accumulation in adipose tissue and other organs, such as skeletal muscle, whereas aerobic exercise (AE) plays an important role in managing obesity through profound protein regulation. Our study aimed to investigate the impact of AE on proteomic changes in both the skeletal muscle and the epididymal fat pad (EFP) of high-fat-diet-induced obese mice. Bioinformatic analyses were performed on differentially regulated proteins using gene ontology enrichment analysis and ingenuity pathway analysis. Eight weeks of AE significantly reduced body weight, increased the serum FNDC5 level, and improved the homeostatic model assessment of insulin resistance. A high-fat diet caused alterations in a subset of proteins involved in the sirtuin signaling pathway and the production of reactive oxygen species in both skeletal muscle and EFP, leading to insulin resistance, mitochondrial dysfunction, and inflammation. On the other hand, AE upregulated skeletal muscle proteins (NDUFB5, NDUFS2, NDUFS7, ETFD, FRDA, and MKNK1) that enhance mitochondrial function and insulin sensitivity. Additionally, the upregulation of LDHC and PRKACA and the downregulation of CTBP1 in EFP can promote the browning of white adipose tissue with the involvement of FNDC5/irisin in the canonical pathway. Our study provides insights into AE-induced molecular responses and may help further develop exercise-mimicking therapeutic targets.
Collapse
|
22
|
Lapatto HA, Kuusela M, Heikkinen A, Muniandy M, van der Kolk BW, Gopalakrishnan S, Pöllänen N, Sandvik M, Schmidt MS, Heinonen S, Saari S, Kuula J, Hakkarainen A, Tampio J, Saarinen T, Taskinen MR, Lundbom N, Groop PH, Tiirola M, Katajisto P, Lehtonen M, Brenner C, Kaprio J, Pekkala S, Ollikainen M, Pietiläinen KH, Pirinen E. Nicotinamide riboside improves muscle mitochondrial biogenesis, satellite cell differentiation, and gut microbiota in a twin study. SCIENCE ADVANCES 2023; 9:eadd5163. [PMID: 36638183 PMCID: PMC9839336 DOI: 10.1126/sciadv.add5163] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) precursor nicotinamide riboside (NR) has emerged as a promising compound to improve obesity-associated mitochondrial dysfunction and metabolic syndrome in mice. However, most short-term clinical trials conducted so far have not reported positive outcomes. Therefore, we aimed to determine whether long-term NR supplementation boosts mitochondrial biogenesis and metabolic health in humans. Twenty body mass index (BMI)-discordant monozygotic twin pairs were supplemented with an escalating dose of NR (250 to 1000 mg/day) for 5 months. NR improved systemic NAD+ metabolism, muscle mitochondrial number, myoblast differentiation, and gut microbiota composition in both cotwins. NR also showed a capacity to modulate epigenetic control of gene expression in muscle and adipose tissue in both cotwins. However, NR did not ameliorate adiposity or metabolic health. Overall, our results suggest that NR acts as a potent modifier of NAD+ metabolism, muscle mitochondrial biogenesis and stem cell function, gut microbiota, and DNA methylation in humans irrespective of BMI.
Collapse
Affiliation(s)
- Helena A. K. Lapatto
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Minna Kuusela
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Aino Heikkinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Maheswary Muniandy
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Birgitta W. van der Kolk
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | | | - Noora Pöllänen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Martin Sandvik
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mark S. Schmidt
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Sina Saari
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Juho Kuula
- Department of Radiology, Medical Imaging Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
- Population Health Unit, Finnish Institute for Health and Welfare, Oulu, Finland
| | - Antti Hakkarainen
- Department of Radiology, Medical Imaging Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Janne Tampio
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Tuure Saarinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
- Abdominal Center, Department of Gastrointestinal Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Marja-Riitta Taskinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Nina Lundbom
- Department of Radiology, Medical Imaging Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center, Nephrology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Marja Tiirola
- Department of Environmental and Biological Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland
| | - Pekka Katajisto
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Charles Brenner
- Department of Diabetes and Cancer Metabolism, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jaakko Kaprio
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Satu Pekkala
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland
| | - Miina Ollikainen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Kirsi H. Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
- Abdominal Center, Healthy Weight Hub, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
- Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, FIN-90220 Oulu, Finland
| |
Collapse
|
23
|
García-Pérez R, Ramirez JM, Ripoll-Cladellas A, Chazarra-Gil R, Oliveros W, Soldatkina O, Bosio M, Rognon PJ, Capella-Gutierrez S, Calvo M, Reverter F, Guigó R, Aguet F, Ferreira PG, Ardlie KG, Melé M. The landscape of expression and alternative splicing variation across human traits. CELL GENOMICS 2023; 3:100244. [PMID: 36777183 PMCID: PMC9903719 DOI: 10.1016/j.xgen.2022.100244] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/08/2022] [Accepted: 12/07/2022] [Indexed: 12/31/2022]
Abstract
Understanding the consequences of individual transcriptome variation is fundamental to deciphering human biology and disease. We implement a statistical framework to quantify the contributions of 21 individual traits as drivers of gene expression and alternative splicing variation across 46 human tissues and 781 individuals from the Genotype-Tissue Expression project. We demonstrate that ancestry, sex, age, and BMI make additive and tissue-specific contributions to expression variability, whereas interactions are rare. Variation in splicing is dominated by ancestry and is under genetic control in most tissues, with ribosomal proteins showing a strong enrichment of tissue-shared splicing events. Our analyses reveal a systemic contribution of types 1 and 2 diabetes to tissue transcriptome variation with the strongest signal in the nerve, where histopathology image analysis identifies novel genes related to diabetic neuropathy. Our multi-tissue and multi-trait approach provides an extensive characterization of the main drivers of human transcriptome variation in health and disease.
Collapse
Affiliation(s)
- Raquel García-Pérez
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Jose Miguel Ramirez
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Aida Ripoll-Cladellas
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Ruben Chazarra-Gil
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Winona Oliveros
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Oleksandra Soldatkina
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Mattia Bosio
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Paul Joris Rognon
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
- Department of Economics and Business, Universitat Pompeu Fabra, Barcelona, Catalonia 08005, Spain
- Department of Statistics and Operations Research, Universitat Politècnica de Catalunya, Barcelona, Catalonia 08034, Spain
| | - Salvador Capella-Gutierrez
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Miquel Calvo
- Statistics Section, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Catalonia 08028, Spain
| | - Ferran Reverter
- Statistics Section, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Catalonia 08028, Spain
| | - Roderic Guigó
- Bioinformatics and Genomics, Center for Genomic Regulation, Barcelona, Catalonia 08003, Spain
| | | | - Pedro G. Ferreira
- Department of Computer Science, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal
- Laboratory of Artificial Intelligence and Decision Support, INESC TEC, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto, Institute for Research and Innovation in Health (i3s), R. Alfredo Allen 208, 4200-135 Porto, Portugal
| | | | - Marta Melé
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| |
Collapse
|
24
|
Nogueira AVB, Lopes MES, Marcantonio CC, Salmon CR, Mofatto LS, Deschner J, Nociti-Junior FH, Cirelli JA. Obesity Modifies the Proteomic Profile of the Periodontal Ligament. Int J Mol Sci 2023; 24:ijms24021003. [PMID: 36674516 PMCID: PMC9861657 DOI: 10.3390/ijms24021003] [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: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023] Open
Abstract
This study aimed to assess the obesity effects on the proteomic profile of the periodontal ligament of rats submitted to obesity induction by a high-fat diet. Eight Holtzman rats were divided into control (n = 3) and obese (n = 5) groups. The maxillae were histologically processed for laser capture microdissection of the periodontal ligament of the first maxillary molars. Peptide mixtures were analyzed by LC-MS/MS. A total of 1379 proteins were identified in all groups. Among them, 335 (24.30%) were exclusively detected in the obese group, while 129 (9.35%) proteins were uniquely found in the control group. Out of the 110 (7.98%) differentially abundant proteins, 10 were more abundant and 100 had decreased abundance in the obese group. A gene ontology analysis showed some proteins related to obesity in the “extracellular exosome” term among differentially identified proteins in the gene ontology cellular component terms Prelp, Sec13, and Sod2. These three proteins were upregulated in the obese group (p < 0.05), as shown by proteomic and immunohistochemistry analyses. In summary, our study presents novel evidence that the proteomic profile of the periodontal ligament is altered in experimental obesity induction, providing a list of differentially abundant proteins associated with obesity, which indicates that the periodontal ligament is responsive to obesity.
Collapse
Affiliation(s)
- Andressa V. B. Nogueira
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University—UNESP, Araraquara 14801-903, São Paulo, Brazil
- Correspondence: (A.V.B.N.); (J.A.C.); Tel.: +49-0-6131-17-7091 (A.V.B.N.); +55-16-3301-6375 (J.A.C.)
| | - Maria Eduarda S. Lopes
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University—UNESP, Araraquara 14801-903, São Paulo, Brazil
| | - Camila C. Marcantonio
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University—UNESP, Araraquara 14801-903, São Paulo, Brazil
| | - Cristiane R. Salmon
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas—UNICAMP, Piracicaba 13414-903, São Paulo, Brazil
| | - Luciana S. Mofatto
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas—UNICAMP, Campinas 13083-862, São Paulo, Brazil
| | - James Deschner
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Francisco H. Nociti-Junior
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas—UNICAMP, Piracicaba 13414-903, São Paulo, Brazil
- São Leopoldo Mandic Research Center, Campinas 13045-755, São Paulo, Brazil
| | - Joni A. Cirelli
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University—UNESP, Araraquara 14801-903, São Paulo, Brazil
- Correspondence: (A.V.B.N.); (J.A.C.); Tel.: +49-0-6131-17-7091 (A.V.B.N.); +55-16-3301-6375 (J.A.C.)
| |
Collapse
|
25
|
Aragón-Vela J, Alcalá-Bejarano Carrillo J, Moreno-Racero A, Plaza-Diaz J. The Role of Molecular and Hormonal Factors in Obesity and the Effects of Physical Activity in Children. Int J Mol Sci 2022; 23:15413. [PMID: 36499740 PMCID: PMC9737554 DOI: 10.3390/ijms232315413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/27/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Obesity and overweight are defined as abnormal fat accumulations. Adipose tissue consists of more than merely adipocytes; each adipocyte is closely coupled with the extracellular matrix. Adipose tissue stores excess energy through expansion. Obesity is caused by the abnormal expansion of adipose tissue as a result of adipocyte hypertrophy and hyperplasia. The process of obesity is controlled by several molecules, such as integrins, kindlins, or matrix metalloproteinases. In children with obesity, metabolomics studies have provided insight into the existence of unique metabolic profiles. As a result of low-grade inflammation in the system, abnormalities were observed in several metabolites associated with lipid, carbohydrate, and amino acid pathways. In addition, obesity and related hormones, such as leptin, play an instrumental role in regulating food intake and contributing to childhood obesity. The World Health Organization states that physical activity benefits the heart, the body, and the mind. Several noncommunicable diseases, such as cardiovascular disease, cancer, and diabetes, can be prevented and managed through physical activity. In this work, we reviewed pediatric studies that examined the molecular and hormonal control of obesity and the influence of physical activity on children with obesity or overweight. The purpose of this review was to examine some orchestrators involved in this disease and how they are related to pediatric populations. A larger number of randomized clinical trials with larger sample sizes and long-term studies could lead to the discovery of new key molecules as well as the detection of significant factors in the coming years. In order to improve the health of the pediatric population, omics analyses and machine learning techniques can be combined in order to improve treatment decisions.
Collapse
Affiliation(s)
- Jerónimo Aragón-Vela
- Department of Health Sciences, Area of Physiology, Building B3, Campus s/n “Las Lagunillas”, University of Jaén, 23071 Jaén, Spain
| | - Jesús Alcalá-Bejarano Carrillo
- Department of Health, University of the Valley of Mexico, Robles 600, Tecnologico I, San Luis Potosí 78220, Mexico
- Research and Advances in Molecular and Cellular Immunology, Center of Biomedical Research, University of Granada, Avda, del Conocimiento s/n, 18016 Armilla, Spain
| | - Aurora Moreno-Racero
- Research and Advances in Molecular and Cellular Immunology, Center of Biomedical Research, University of Granada, Avda, del Conocimiento s/n, 18016 Armilla, Spain
| | - Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Instituto de Investigación Biosanitaria IBS, Granada, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
| |
Collapse
|
26
|
Garske KM, Comenho C, Pan DZ, Alvarez M, Mohlke K, Laakso M, Pietiläinen KH, Pajukanta P. Long-range chromosomal interactions increase and mark repressed gene expression during adipogenesis. Epigenetics 2022; 17:1849-1862. [PMID: 35746833 PMCID: PMC9665133 DOI: 10.1080/15592294.2022.2088145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Obesity perturbs central functions of human adipose tissue, centred on differentiation of preadipocytes to adipocytes, i.e., adipogenesis. The large environmental component of obesity makes it important to elucidate epigenetic regulatory factors impacting adipogenesis. Promoter Capture Hi-C (pCHi-C) has been used to identify chromosomal interactions between promoters and associated regulatory elements. However, long range interactions (LRIs) greater than 1 Mb are often filtered out of pCHi-C datasets, due to technical challenges and their low prevalence. To elucidate the unknown role of LRIs in adipogenesis, we investigated preadipocyte differentiation to adipocytes using pCHi-C and bulk and single nucleus RNA-seq data. We first show that LRIs are reproducible between biological replicates, and they increase >2-fold in frequency across adipogenesis. We further demonstrate that genomic loci containing LRIs are more epigenetically repressed than regions without LRIs, corresponding to lower gene expression in the LRI regions. Accordingly, as preadipocytes differentiate into adipocytes, LRI regions are more likely to contain repressed preadipocyte marker genes; whereas these same LRI regions are depleted of actively expressed adipocyte marker genes. Finally, we show that LRIs can be used to restrict multiple testing of the long-range cis-eQTL analysis to identify variants that regulate genes via LRIs. We exemplify this by identifying a putative long range cis regulatory mechanism at the LYPLAL1/TGFB2 obesity locus. In summary, we identify LRIs that mark repressed regions of the genome, and these interactions increase across adipogenesis, pinpointing developmental regions that need to be repressed in a cell-type specific way for adipogenesis to proceed.
Collapse
Affiliation(s)
- Kristina M. Garske
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Caroline Comenho
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - David Z. Pan
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA,Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Karen Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Markku Laakso
- Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Kirsi H. Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland,Obesity Center, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA,Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA,Institute for Precision Heath, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA,CONTACT Päivi Pajukanta Department of Human Genetics David Geffen School of Medicine at UCLA
| |
Collapse
|
27
|
Wilson MR, Skalski H, Reske JJ, Wegener M, Adams M, Hostetter G, Hoffmann HM, Bernard JJ, Bae-Jump VL, Teixeira JM, Chandler RL. Obesity alters the mouse endometrial transcriptome in a cell context-dependent manner. Reprod Biol Endocrinol 2022; 20:163. [PMID: 36424602 PMCID: PMC9686036 DOI: 10.1186/s12958-022-01030-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2022] Open
Abstract
Obesity impacts fertility and is positively correlated with endometrial hyperplasia and endometrial cancer occurrence. Endometrial epithelia often harbor disease driver-mutations, while endometrial stroma are highly regulative of neighboring epithelia. Here, we sought to determine distinct transcriptome changes occurring in individual cell types in the obese mouse uterus. Outbred CD-1 mice were fed high-fat or control diets for 18 weeks, estrous cycle staged, and endometrial epithelia, macrophages, and stroma isolated for transcriptomic analysis. High-fat diet mice displayed increased body mass and developed glucose intolerance, hyperinsulinemia, and fatty liver. Obese mouse epithelia displayed differential gene expression for genes related to innate immunity and leukocyte chemotaxis. The obese mouse stroma differentially expressed factors related to circadian rhythm, and expression of these genes correlated with glucose tolerance or body mass. We observed correlations between F4/80 + macrophage numbers, Cleaved Caspase 3 (CC3) apoptosis marker staining and glucose intolerance among obese mice, including a subgroup of obese mice with high CC3 + luminal epithelia. This subgroup displayed differential gene expression among all cell types, with pathways related to immune escape in epithelia and macrophages, while the stroma dysregulated pathways related to regulation of epithelia. These results suggest an important role for differential response of both the epithelia and stroma in their response to obesity, while macrophages are dysregulated in the context of apoptotic epithelia. The obesity-related gene expression programs in cells within the uterine microenvironment may influence the ability of the endometrium to function during pregnancy and influence disease pathogenesis.
Collapse
Affiliation(s)
- Mike R Wilson
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Hilary Skalski
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Jake J Reske
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Marc Wegener
- Genomics Core Facility, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Marie Adams
- Genomics Core Facility, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Galen Hostetter
- Pathology and Biorepository Core, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Hanne M Hoffmann
- Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, 48824, USA
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Jamie J Bernard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, 48824, USA
- Division of Dermatology, Department of Medicine, Michigan State University, East Lansing, MI, USA
| | - Victoria L Bae-Jump
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jose M Teixeira
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
- Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Ronald L Chandler
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA.
- Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
28
|
Coletta S, Trevellin E, Benagiano M, Romagnoli J, Della Bella C, D’Elios MM, Vettor R, de Bernard M. The antigen CD300e drives T cell inflammation in adipose tissue and elicits an antibody response predictive of the insulin sensitivity recovery in obese patients. J Inflamm (Lond) 2022; 19:21. [PMID: 36419085 PMCID: PMC9682732 DOI: 10.1186/s12950-022-00318-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/04/2022] [Indexed: 11/24/2022] Open
Abstract
Obesity and insulin resistance (IR), the key features of metabolic syndrome, are closely associated with a state of chronic, low-grade inflammation. Bariatric surgery leads to a considerable reduction in the adipose tissue mass and systemic inflammation along with a reduction of IR, with a whole-body metabolic improvement. However, a sizable portion of people experience an IR relapse within few years of remission.Numerous studies have attempted to explore the best clinical predictors of the improvement of insulin sensitivity and the maintenance of glucose homeostasis after bariatric surgery, but no simple fasting blood test has been found to be effective in predicting the short and long-term beneficial effects on glycaemia.With the present study, we investigated T-cell and antibody responses against CD300e, an antigen highly expressed in the adipose tissue of patients with obesity before the bariatric surgery-induced weight loss. We found both in fat tissue and in peripheral blood anti-CD300e-specific T helper 1 responses. Moreover, we evidenced in the sera of individuals with obesity an antibody response towards CD300e and revealed the existence of a significant correlation between the level of antibodies before surgery and the maintenance of glucose control after the intervention.
Collapse
Affiliation(s)
- Sara Coletta
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padova, Padova, Italy
| | - Elisabetta Trevellin
- grid.5608.b0000 0004 1757 3470Medical Clinic III, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Marisa Benagiano
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Jacopo Romagnoli
- grid.8142.f0000 0001 0941 3192Department of Medicine and Translational Surgery, Catholic University, Rome, Italy
| | - Chiara Della Bella
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mario Milco D’Elios
- grid.9024.f0000 0004 1757 4641Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Roberto Vettor
- grid.5608.b0000 0004 1757 3470Medical Clinic III, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Marina de Bernard
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
29
|
Wu LD, Kong CH, Shi Y, Zhang JX, Chen SL. Associations between novel anthropometric measures and the prevalence of hypertension among 45,853 adults: A cross-sectional study. Front Cardiovasc Med 2022; 9:1050654. [PMID: 36407444 PMCID: PMC9669705 DOI: 10.3389/fcvm.2022.1050654] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/19/2022] [Indexed: 02/03/2024] Open
Abstract
AIMS Traditional anthropometric measures, including body mass index (BMI), are insufficient for evaluating the risk of hypertension. We aimed to investigate the association between novel anthropometric indices and hypertension risk in a large population in the United States. METHODS Forty-five thousand eight hundred fifty-three participants from the National Health and Nutrition Examination Survey (NHANES) (1999-2018) were enrolled. Social demographic information, lifestyle factors, blood biochemical measurements and anthropometric indices, including body weight, body mass index (BMI), waist circumference, waist-to-height ratio (WtHR), conicity index (CI), a body shape index (ABSI), body roundness index (BRI) and lipid accumulation product (LAP) were collected. Multivariable logistic regression and restricted cubic spline were adopted to investigate the associations between hypertension risk and anthropometric indices. We also performed receiver operating characteristic (ROC) curve analyses to further evaluate the discriminatory powers of anthropometric measurements for screening hypertension risk. Moreover, participants were randomly assigned to the training group and the validation group in a ratio of 3 to 1. A nomogram model based on anthropometric measures was established and validated in the training group and validation group, respectively. RESULTS All of the anthropometric measurements investigated were positively and independently associated with the hypertension risk. Among all anthropometric indices, per-SD increment in ABSI had the highest OR (OR: 3.4; 95% CI: 2.73-4.24) after adjusting for age, sex, race/ethnicity, education, smoking, drinking, diabetes, and eGFR. Moreover, results from restricted cubic splines revealed the non-linear association between anthropometric measurements and hypertension risk. In ROC analyses, CI had superior discriminatory power for hypertension (area under the curve: 0.71; 95% CI: 0.706-0.715; optimal cutoff value: 1.3) compared with other indices. Nomogram model based on age, sex, diabetes, CI and LAP showed favorable predicting ability of hypertension risk with an AUC (95% CI) in training group of 80.2% (79.7-80.6%), and the AUC (95% CI) in validation group was 79.5% (78.3-80.1%). Meanwhile, calibration plot showed good consistency. CONCLUSIONS Anthropometric measurements including BMI, WtHR, CI, ABSI, BRI and LAP are closely associated with hypertension risk in the present study. For better prevention and treatment of hypertension, more attention should be paid to anthropometric indices, especially novel anthropometric indices.
Collapse
Affiliation(s)
| | | | | | - Jun-Xia Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shao-Liang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
30
|
Effects of Whey Protein or Its Hydrolysate Supplements Combined with an Energy-Restricted Diet on Weight Loss: A Randomized Controlled Trial in Older Women. Nutrients 2022; 14:nu14214540. [PMID: 36364801 PMCID: PMC9657015 DOI: 10.3390/nu14214540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/03/2022] [Accepted: 10/25/2022] [Indexed: 12/03/2022] Open
Abstract
An energy-restricted weight-loss approach has limitations when it used in the elderly, especially because of muscle loss. We aimed to assess the effects of whey protein (WP) or WP hydrolysate (WPH) combined with an energy-restricted diet (ERD) on weight reduction and muscle preservation in older women with overweight and obesity. A total of 60 women were randomized to the control (ERD), WP (ERD + 20 g/d WP) or WPH (ERD + 20 g/d WPH) group, using a 1:1:1 allocation ratio. After an 8-week intervention, body composition, gut microbiota, and serum metabolomics changes were compared among the three groups. The reductions in body weight (−1.11 ± 1.11 vs. −2.34 ± 1.35, p < 0.05), BMI (−0.46 ± 0.45 vs. −0.97 ± 0.54, p < 0.05), and body fat (−0.70 ± 0.92 vs. −2.45 ± 1.65, p < 0.01) were higher in the WPH group than in the control group. Body fat (%) was significantly decreased in the two protein groups. Fat-free mass did not significantly change among the three groups. Serum metabolomics showed that the tricarboxylic acid cycle pathway was upregulated in the WPH group. No significant changes in microbiota were observed among the groups. In conclusion, WP or WPH supplementation combined with an energy-restricted diet benefits older women during weight loss. WPH was more effective, possibly due to increased energy metabolism.
Collapse
|
31
|
Fisk HL, Childs CE, Miles EA, Ayres R, Noakes PS, Paras-Chavez C, Antoun E, Lillycrop KA, Calder PC. Dysregulation of Subcutaneous White Adipose Tissue Inflammatory Environment Modelling in Non-Insulin Resistant Obesity and Responses to Omega-3 Fatty Acids – A Double Blind, Randomised Clinical Trial. Front Immunol 2022; 13:922654. [PMID: 35958557 PMCID: PMC9358040 DOI: 10.3389/fimmu.2022.922654] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/20/2022] [Indexed: 01/15/2023] Open
Abstract
Background Obesity is associated with enhanced lipid accumulation and the expansion of adipose tissue accompanied by hypoxia and inflammatory signalling. Investigation in human subcutaneous white adipose tissue (scWAT) in people living with obesity in which metabolic complications such as insulin resistance are yet to manifest is limited, and the mechanisms by which these processes are dysregulated are not well elucidated. Long chain omega-3 polyunsaturated fatty acids (LC n-3 PUFAs) have been shown to modulate the expression of genes associated with lipid accumulation and collagen deposition and reduce the number of inflammatory macrophages in adipose tissue from individuals with insulin resistance. Therefore, these lipids may have positive actions on obesity associated scWAT hypertrophy and inflammation. Methods To evaluate obesity-associated tissue remodelling and responses to LC n-3 PUFAs, abdominal scWAT biopsies were collected from normal weight individuals and those living with obesity prior to and following 12-week intervention with marine LC n-3 PUFAs (1.1 g EPA + 0.8 g DHA daily). RNA sequencing, qRT-PCR, and histochemical staining were used to assess remodelling- and inflammatory-associated gene expression, tissue morphology and macrophage infiltration. Results Obesity was associated with scWAT hypertrophy (P < 0.001), hypoxia, remodelling, and inflammatory macrophage infiltration (P = 0.023). Furthermore, we highlight the novel dysregulation of Wnt signalling in scWAT in non-insulin resistant obesity. LC n-3 PUFAs beneficially modulated the scWAT environment through downregulating the expression of genes associated with inflammatory and remodelling pathways (P <0.001), but there were altered outcomes in individuals living with obesity in comparison to normal weight individuals. Conclusion Our data identify dysregulation of Wnt signalling, hypoxia, and hypertrophy, and enhanced macrophage infiltration in scWAT in non-insulin resistant obesity. LC n-3 PUFAs modulate some of these processes, especially in normal weight individuals which may be preventative and limit the development of restrictive and inflammatory scWAT in the development of obesity. We conclude that a higher dose or longer duration of LC n-3 PUFA intervention may be needed to reduce obesity-associated scWAT inflammation and promote tissue homeostasis. Clinical Trial Registration www.isrctn.com, identifier ISRCTN96712688.
Collapse
Affiliation(s)
- Helena L Fisk
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Caroline E Childs
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Elizabeth A Miles
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Robert Ayres
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Paul S Noakes
- School of Medicine, The University of Notre Dame Australia, Freemantle, WA, Australia
| | | | - Elie Antoun
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Karen A Lillycrop
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Philip C Calder
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- National Institute for Health and Care Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton National Health Service (NHS) Foundation Trust and University of Southampton, Southampton, United Kingdom
| |
Collapse
|
32
|
Azzolini F, Berentsen GD, Skaug HJ, Hjelmborg JVB, Kaprio JA. The heritability of BMI varies across the range of BMI-a heritability curve analysis in a twin cohort. Int J Obes (Lond) 2022; 46:1786-1791. [PMID: 35817850 DOI: 10.1038/s41366-022-01172-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND The heritability of traits such as body mass index (BMI), a measure of obesity, is generally estimated using family and twin studies, and increasingly by molecular genetic approaches. These studies generally assume that genetic effects are uniform across all trait values, yet there is emerging evidence that this may not always be the case. METHOD/SUBJECTS This paper analyzes twin data using a recently developed measure of heritability called the heritability curve. Under the assumption that trait values in twin pairs are governed by a flexible Gaussian mixture distribution, heritability curves may vary across trait values. The data consist of repeated measures of BMI on 1506 monozygotic (MZ) and 2843 like-sexed dizygotic (DZ) adult twin pairs, gathered from multiple surveys in older Finnish Twin Cohorts. RESULTS The heritability curve and BMI value-specific MZ and DZ pairwise correlations were estimated, and these varied across the range of BMI. MZ correlations were highest at BMI values from 21 to 24, with a stronger decrease for women than for men at higher values. Models with additive and dominance effects fit best at low and high BMI values, while models with additive genetic and common environmental effects fit best in the normal range of BMI. CONCLUSIONS We demonstrate that twin and molecular genetic studies need to consider how genetic effects vary across trait values. Such variation may reconcile findings of traits with high heritability and major differences in mean values between countries or over time.
Collapse
Affiliation(s)
| | - Geir D Berentsen
- Department of Business and Management Science, NHH Norwegian School of Economics, Bergen, Norway
| | - Hans J Skaug
- Department of Mathematics, University of Bergen, Bergen, Norway
| | - Jacob V B Hjelmborg
- Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jaakko A Kaprio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| |
Collapse
|
33
|
Noble AJ, Purcell RV, Adams AT, Lam YK, Ring PM, Anderson JR, Osborne AJ. A Final Frontier in Environment-Genome Interactions? Integrated, Multi-Omic Approaches to Predictions of Non-Communicable Disease Risk. Front Genet 2022; 13:831866. [PMID: 35211161 PMCID: PMC8861380 DOI: 10.3389/fgene.2022.831866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/19/2022] [Indexed: 12/26/2022] Open
Abstract
Epidemiological and associative research from humans and animals identifies correlations between the environment and health impacts. The environment-health inter-relationship is effected through an individual's underlying genetic variation and mediated by mechanisms that include the changes to gene regulation that are associated with the diversity of phenotypes we exhibit. However, the causal relationships have yet to be established, in part because the associations are reduced to individual interactions and the combinatorial effects are rarely studied. This problem is exacerbated by the fact that our genomes are highly dynamic; they integrate information across multiple levels (from linear sequence, to structural organisation, to temporal variation) each of which is open to and responds to environmental influence. To unravel the complexities of the genomic basis of human disease, and in particular non-communicable diseases that are also influenced by the environment (e.g., obesity, type II diabetes, cancer, multiple sclerosis, some neurodegenerative diseases, inflammatory bowel disease, rheumatoid arthritis) it is imperative that we fully integrate multiple layers of genomic data. Here we review current progress in integrated genomic data analysis, and discuss cases where data integration would lead to significant advances in our ability to predict how the environment may impact on our health. We also outline limitations which should form the basis of future research questions. In so doing, this review will lay the foundations for future research into the impact of the environment on our health.
Collapse
Affiliation(s)
- Alexandra J. Noble
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom
| | - Rachel V. Purcell
- Department of Surgery, University of Otago Christchurch, Christchurch, New Zealand
| | - Alex T. Adams
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom
| | - Ying K. Lam
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom
| | - Paulina M. Ring
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Jessica R. Anderson
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Amy J. Osborne
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| |
Collapse
|
34
|
Patil AD, Aphale PS, Sharma DB, Bhonde RR. Can homeopathic medicine Chelidonium majus serve a dual role of an anti-obesity and anti-diabetic agent? Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2021.110749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
35
|
Hernandez-Castillo C, Shuck SC. Diet and Obesity-Induced Methylglyoxal Production and Links to Metabolic Disease. Chem Res Toxicol 2021; 34:2424-2440. [PMID: 34851609 DOI: 10.1021/acs.chemrestox.1c00221] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The obesity rate in the United States is 42.4% and has become a national epidemic. Obesity is a complex condition that is influenced by socioeconomic status, ethnicity, genetics, age, and diet. Increased consumption of a Western diet, one that is high in processed foods, red meat, and sugar content, is associated with elevated obesity rates. Factors that increase obesity risk, such as socioeconomic status, also increase consumption of a Western diet because of a limited access to healthier options and greater affordability of processed foods. Obesity is a public health threat because it increases the risk of several pathologies, including atherosclerosis, diabetes, and cancer. The molecular mechanisms linking obesity to disease onset and progression are not well understood, but a proposed mechanism is physiological changes caused by altered lipid peroxidation, glycolysis, and protein metabolism. These metabolic pathways give rise to reactive molecules such as the abundant electrophile methylglyoxal (MG), which covalently modifies nucleic acids and proteins. MG-adducts are associated with obesity-linked pathologies and may have potential for biomonitoring to determine the risk of disease onset and progression. MG-adducts may also play a role in disease progression because they are mutagenic and directly impact protein stability and function. In this review, we discuss how obesity drives metabolic alterations, how these alterations lead to MG production, the association of MG-adducts with disease, and the potential impact of MG-adducts on cellular function.
Collapse
Affiliation(s)
- Carlos Hernandez-Castillo
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010, United States
| | - Sarah C Shuck
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010, United States
| |
Collapse
|
36
|
Rossmeislová L, Gojda J, Smolková K. Pancreatic cancer: branched-chain amino acids as putative key metabolic regulators? Cancer Metastasis Rev 2021; 40:1115-1139. [PMID: 34962613 DOI: 10.1007/s10555-021-10016-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/18/2021] [Indexed: 02/06/2023]
Abstract
Branched-chain amino acids (BCAA) are essential amino acids utilized in anabolic and catabolic metabolism. While extensively studied in obesity and diabetes, recent evidence suggests an important role for BCAA metabolism in cancer. Elevated plasma levels of BCAA are associated with an increased risk of developing pancreatic cancer, namely pancreatic ductal adenocarcinoma (PDAC), a tumor with one of the highest 1-year mortality rates. The dreadful prognosis for PDAC patients could be attributable also to the early and frequent development of cancer cachexia, a fatal host metabolic reprogramming leading to muscle and adipose wasting. We propose that BCAA dysmetabolism is a unifying component of several pathological conditions, i.e., obesity, insulin resistance, and PDAC. These conditions are mutually dependent since PDAC ranks among cancers tightly associated with obesity and insulin resistance. It is also well-established that PDAC itself can trigger insulin resistance and new-onset diabetes. However, the exact link between BCAA metabolism, development of PDAC, and tissue wasting is still unclear. Although tissue-specific intracellular and systemic metabolism of BCAA is being intensively studied, unresolved questions related to PDAC and cancer cachexia remain, namely, whether elevated circulating BCAA contribute to PDAC etiology, what is the biological background of BCAA elevation, and what is the role of adipose tissue relative to BCAA metabolism during cancer cachexia. To cover those issues, we provide our view on BCAA metabolism at the intracellular, tissue, and whole-body level, with special emphasis on different metabolic links to BCAA intermediates and the role of insulin in substrate handling.
Collapse
Affiliation(s)
- Lenka Rossmeislová
- Department of Pathophysiology, Center for Research On Nutrition, Metabolism, and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research On Obesity, Third Faculty of Medicine, Prague, Czech Republic
| | - Jan Gojda
- Franco-Czech Laboratory for Clinical Research On Obesity, Third Faculty of Medicine, Prague, Czech Republic
- Department of Internal Medicine, Královské Vinohrady University Hospital and Third Faculty of Medicine, Prague, Czech Republic
| | - Katarína Smolková
- Laboratory of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
37
|
Pan DZ, Miao Z, Comenho C, Rajkumar S, Koka A, Lee SHT, Alvarez M, Kaminska D, Ko A, Sinsheimer JS, Mohlke KL, Mancuso N, Muñoz-Hernandez LL, Herrera-Hernandez M, Tusié-Luna MT, Aguilar-Salinas C, Pietiläinen KH, Pihlajamäki J, Laakso M, Garske KM, Pajukanta P. Identification of TBX15 as an adipose master trans regulator of abdominal obesity genes. Genome Med 2021; 13:123. [PMID: 34340684 PMCID: PMC8327600 DOI: 10.1186/s13073-021-00939-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 07/14/2021] [Indexed: 12/14/2022] Open
Abstract
Background Obesity predisposes individuals to multiple cardiometabolic disorders, including type 2 diabetes (T2D). As body mass index (BMI) cannot reliably differentiate fat from lean mass, the metabolically detrimental abdominal obesity has been estimated using waist-hip ratio (WHR). Waist-hip ratio adjusted for body mass index (WHRadjBMI) in turn is a well-established sex-specific marker for abdominal fat and adiposity, and a predictor of adverse metabolic outcomes, such as T2D. However, the underlying genes and regulatory mechanisms orchestrating the sex differences in obesity and body fat distribution in humans are not well understood. Methods We searched for genetic master regulators of WHRadjBMI by employing integrative genomics approaches on human subcutaneous adipose RNA sequencing (RNA-seq) data (n ~ 1400) and WHRadjBMI GWAS data (n ~ 700,000) from the WHRadjBMI GWAS cohorts and the UK Biobank (UKB), using co-expression network, transcriptome-wide association study (TWAS), and polygenic risk score (PRS) approaches. Finally, we functionally verified our genomic results using gene knockdown experiments in a human primary cell type that is critical for adipose tissue function. Results Here, we identified an adipose gene co-expression network that contains 35 obesity GWAS genes and explains a significant amount of polygenic risk for abdominal obesity and T2D in the UKB (n = 392,551) in a sex-dependent way. We showed that this network is preserved in the adipose tissue data from the Finnish Kuopio Obesity Study and Mexican Obesity Study. The network is controlled by a novel adipose master transcription factor (TF), TBX15, a WHRadjBMI GWAS gene that regulates the network in trans. Knockdown of TBX15 in human primary preadipocytes resulted in changes in expression of 130 network genes, including the key adipose TFs, PPARG and KLF15, which were significantly impacted (FDR < 0.05), thus functionally verifying the trans regulatory effect of TBX15 on the WHRadjBMI co-expression network. Conclusions Our study discovers a novel key function for the TBX15 TF in trans regulating an adipose co-expression network of 347 adipose, mitochondrial, and metabolically important genes, including PPARG, KLF15, PPARA, ADIPOQ, and 35 obesity GWAS genes. Thus, based on our converging genomic, transcriptional, and functional evidence, we interpret the role of TBX15 to be a main transcriptional regulator in the adipose tissue and discover its importance in human abdominal obesity. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-021-00939-2.
Collapse
Affiliation(s)
- David Z Pan
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA.,Bioinformatics Interdepartmental Program, UCLA, Los Angeles, USA
| | - Zong Miao
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA.,Bioinformatics Interdepartmental Program, UCLA, Los Angeles, USA
| | - Caroline Comenho
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Sandhya Rajkumar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA.,Computational and Systems Biology Interdepartmental Program, UCLA, Los Angeles, USA
| | - Amogha Koka
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Seung Hyuk T Lee
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Dorota Kaminska
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA.,Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Arthur Ko
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Janet S Sinsheimer
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA.,Department of Computational Medicine, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nicholas Mancuso
- Center for Genetic Epidemiology, Department of Preventative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Linda Liliana Muñoz-Hernandez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey, N.L., México, 64710.,Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Departamento de Endocrinología y Metabolismo del Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Miguel Herrera-Hernandez
- Departamento de Cirugía, Instituto Nacional de Ciencias Médicas y Nutrición, Mexico City, Mexico
| | - Maria Teresa Tusié-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto de Investigaciones Biomédicas UNAM/ Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Carlos Aguilar-Salinas
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Departamento de Endocrinología y Metabolismo del Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Obesity Center, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Kristina M Garske
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA. .,Bioinformatics Interdepartmental Program, UCLA, Los Angeles, USA. .,Institute for Precision Health at UCLA, Los Angeles, USA.
| |
Collapse
|
38
|
Tokarz J, Möller G, Artati A, Huber S, Zeigerer A, Blaauw B, Adamski J, Dyar KA. Common Muscle Metabolic Signatures Highlight Arginine and Lysine Metabolism as Potential Therapeutic Targets to Combat Unhealthy Aging. Int J Mol Sci 2021; 22:ijms22157958. [PMID: 34360722 PMCID: PMC8348621 DOI: 10.3390/ijms22157958] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022] Open
Abstract
Biological aging research is expected to reveal modifiable molecular mechanisms that can be harnessed to slow or possibly reverse unhealthy trajectories. However, there is first an urgent need to define consensus molecular markers of healthy and unhealthy aging. Established aging hallmarks are all linked to metabolism, and a ‘rewired’ metabolic circuitry has been shown to accelerate or delay biological aging. To identify metabolic signatures distinguishing healthy from unhealthy aging trajectories, we performed nontargeted metabolomics on skeletal muscles from 2-month-old and 21-month-old mice, and after dietary and lifestyle interventions known to impact biological aging. We hypothesized that common metabolic signatures would highlight specific pathways and processes promoting healthy aging, while revealing the molecular underpinnings of unhealthy aging. Here, we report 50 metabolites that commonly distinguished aging trajectories in all cohorts, including 18 commonly reduced under unhealthy aging and 32 increased. We stratified these metabolites according to known relationships with various aging hallmarks and found the greatest associations with oxidative stress and nutrient sensing. Collectively, our data suggest interventions aimed at maintaining skeletal muscle arginine and lysine may be useful therapeutic strategies to minimize biological aging and maintain skeletal muscle health, function, and regenerative capacity in old age.
Collapse
Affiliation(s)
- Janina Tokarz
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (J.T.); (G.M.); (A.Z.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Gabriele Möller
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (J.T.); (G.M.); (A.Z.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Anna Artati
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (A.A.); (S.H.)
| | - Simone Huber
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (A.A.); (S.H.)
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (J.T.); (G.M.); (A.Z.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Bert Blaauw
- Department of Biomedical Sciences, University of Padova, 35129 Padova, Italy;
- Venetian Institute of Molecular Medicine, 35129 Padova, Italy
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000 Ljubljana, Slovenia
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Kenneth Allen Dyar
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (J.T.); (G.M.); (A.Z.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Correspondence:
| |
Collapse
|