1
|
Kang GS, Kim YE, Oh HR, Jo HJ, Bok S, Jeon YK, Cheon GJ, Roh TY, Chang YT, Park DJ, Ahn GO. Hypoxia-inducible factor-1α-deficient adipose-tissue macrophages produce the heat to mediate lipolysis of white adipose tissue through uncoupling protein-1. Lab Anim Res 2024; 40:37. [PMID: 39473019 PMCID: PMC11523771 DOI: 10.1186/s42826-024-00224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Uncoupling protein 1 (UCP1) is a proton uncoupler located across the mitochondrial membrane generally involved in thermogenesis of brown adipose tissues. Although UCP1 is known to be strongly expressed in brown adipocytes, recent evidence suggest that white adipocytes can also express UCP1 under certain circumstances such as cold- or β-adrenergic receptor-stimulation, allowing them to acquire brown adipocyte-like features thereby becoming 'beige' adipocytes. RESULTS In this study, we report that UCP1 can be expressed in adipose-tissue macrophages (ATM) lacking functional hypoxia-inducible factor-1 (HIF-1) and this does not require cold- nor β-adrenergic receptor activation. By using myeloid-specific Hif-1α knockout (KO) mice, we observed that these mice were protected from diet-induced obesity and exhibited an improved thermogenic tolerance upon cold challenge. ATM isolated from white adipose tissues (WAT) of these mice fed with high fat diet exhibited significantly higher M2-polarization, decreased glycolysis, increased mitochondrial functions and acetyl-CoA levels, along with increased expression of Ucp1, peroxisome proliferator activated receptor-gamma co-activator-1a, and others involved in histone acetylation. Consistent with the increased Ucp1 gene expression, these ATM produced a significant amount of heat mediating lipolysis of co-cultured adipocytes liberating free fatty acid. Treating ATM with acetate, a substrate for acetyl-CoA synthesis was able to boost the heat production in wild-type or Hif-1α-deficient but not UCP1-deficient macrophages, indicating that UCP1 was necessary for the heat production in macrophages. Lastly, we observed a significant inverse correlation between the number of UCP1-expressing ATM in WAT and the body mass index of human individuals. CONCLUSIONS UCP1-expressing ATM produce the heat to mediate lipolysis of adipocytes, indicating that this can be a novel strategy to treat and prevent diet-induced obesity.
Collapse
Affiliation(s)
- Gi-Sue Kang
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Young-Eun Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Ho Rim Oh
- Department of Nuclear Medicine, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - Hye-Ju Jo
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Seoyeon Bok
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Yoon Kyung Jeon
- Department of Pathology, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, College of Medicine, Seoul National University, Seoul, 03080, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea
- College of Medicine, Cancer Research Institute, Seoul National University, Seoul, 03080, Korea
| | - Tae-Young Roh
- Department of Life Sciences, Ewha Womans University, Seoul, 03760, Korea
| | | | - Do Joong Park
- Department of Surgery, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - G-One Ahn
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea.
- College of Medicine, Cancer Research Institute, Seoul National University, Seoul, 03080, Korea.
| |
Collapse
|
2
|
Li G, Meex RCR, Goossens GH. The role of tissue oxygenation in obesity-related cardiometabolic complications. Rev Endocr Metab Disord 2024:10.1007/s11154-024-09910-z. [PMID: 39298040 DOI: 10.1007/s11154-024-09910-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2024] [Indexed: 09/21/2024]
Abstract
Obesity is a complex, multifactorial, chronic disease that acts as a gateway to a range of other diseases. Evidence from recent studies suggests that changes in oxygen availability in the microenvironment of metabolic organs may exert an important role in the development of obesity-related cardiometabolic complications. In this review, we will first discuss results from observational and controlled laboratory studies that examined the relationship between reduced oxygen availability and obesity-related metabolic derangements. Next, the effects of alterations in oxygen partial pressure (pO2) in the adipose tissue, skeletal muscle and the liver microenvironment on physiological processes in these key metabolic organs will be addressed, and how this might relate to cardiometabolic complications. Since many obesity-related chronic diseases, including type 2 diabetes mellitus, cardiovascular diseases, chronic kidney disease, chronic obstructive pulmonary disease and obstructive sleep apnea, are characterized by changes in pO2 in the tissue microenvironment, a better understanding of the metabolic impact of altered tissue oxygenation can provide valuable insights into the complex interplay between environmental and biological factors involved in the pathophysiology of metabolic impairments. This may ultimately contribute to the development of novel strategies to prevent and treat obesity-related cardiometabolic diseases.
Collapse
Affiliation(s)
- Geng Li
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands
| | - Ruth C R Meex
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands
| | - Gijs H Goossens
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands.
| |
Collapse
|
3
|
Rodriguez-Muñoz A, Motahari-Rad H, Martin-Chaves L, Benitez-Porres J, Rodriguez-Capitan J, Gonzalez-Jimenez A, Insenser M, Tinahones FJ, Murri M. A Systematic Review of Proteomics in Obesity: Unpacking the Molecular Puzzle. Curr Obes Rep 2024; 13:403-438. [PMID: 38703299 PMCID: PMC11306592 DOI: 10.1007/s13679-024-00561-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/14/2024] [Indexed: 05/06/2024]
Abstract
PURPOSE OF REVIEW The present study aims to review the existing literature to identify pathophysiological proteins in obesity by conducting a systematic review of proteomics studies. Proteomics may reveal the mechanisms of obesity development and clarify the links between obesity and related diseases, improving our comprehension of obesity and its clinical implications. RECENT FINDINGS Most of the molecular events implicated in obesity development remain incomplete. Proteomics stands as a powerful tool for elucidating the intricate interactions among proteins in the context of obesity. This methodology has the potential to identify proteins involved in pathological processes and to evaluate changes in protein abundance during obesity development, contributing to the identification of early disease predisposition, monitoring the effectiveness of interventions and improving disease management overall. Despite many non-targeted proteomic studies exploring obesity, a comprehensive and up-to-date systematic review of the molecular events implicated in obesity development is lacking. The lack of such a review presents a significant challenge for researchers trying to interpret the existing literature. This systematic review was conducted following the PRISMA guidelines and included sixteen human proteomic studies, each of which delineated proteins exhibiting significant alterations in obesity. A total of 41 proteins were reported to be altered in obesity by at least two or more studies. These proteins were involved in metabolic pathways, oxidative stress responses, inflammatory processes, protein folding, coagulation, as well as structure/cytoskeleton. Many of the identified proteomic biomarkers of obesity have also been reported to be dysregulated in obesity-related disease. Among them, seven proteins, which belong to metabolic pathways (aldehyde dehydrogenase and apolipoprotein A1), the chaperone family (albumin, heat shock protein beta 1, protein disulfide-isomerase A3) and oxidative stress and inflammation proteins (catalase and complement C3), could potentially serve as biomarkers for the progression of obesity and the development of comorbidities, contributing to personalized medicine in the field of obesity. Our systematic review in proteomics represents a substantial step forward in unravelling the complexities of protein alterations associated with obesity. It provides valuable insights into the pathophysiological mechanisms underlying obesity, thereby opening avenues for the discovery of potential biomarkers and the development of personalized medicine in obesity.
Collapse
Affiliation(s)
- Alba Rodriguez-Muñoz
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain
| | - Hanieh Motahari-Rad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Laura Martin-Chaves
- Heart Area, Hospital Universitario Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Javier Benitez-Porres
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- Department of Human Physiology, Physical Education and Sport, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Jorge Rodriguez-Capitan
- Heart Area, Hospital Universitario Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Biomedical Research Network Center for Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | | | - Maria Insenser
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology & Nutrition, Hospital Universitario Ramón y Cajal & Universidad de Alcalá & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| | - Francisco J Tinahones
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Mora Murri
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain.
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain.
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
4
|
Han Y, Ye S, Liu B. Roles of extracellular vesicles derived from healthy and obese adipose tissue in inter-organ crosstalk and potential clinical implication. Front Endocrinol (Lausanne) 2024; 15:1409000. [PMID: 39268243 PMCID: PMC11390393 DOI: 10.3389/fendo.2024.1409000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Extracellular vesicles (EVs) are nanovesicles containing bioactive molecules including proteins, nucleic acids and lipids that mediate intercellular and inter-organ communications, holding promise as potential therapeutics for multiple diseases. Adipose tissue (AT) serves as a dynamically distributed energy storage organ throughout the body, whose accumulation leads to obesity, a condition characterized by infiltration with abundant immune cells. Emerging evidence has illustrated that EVs secreted by AT are the novel class of adipokines that regulate the homeostasis between AT and peripheral organs. However, most of the studies focused on the investigations of EVs derived from adipocytes or adipose-derived stem cells (ADSCs), the summarization of functions in cellular and inter-organ crosstalk of EVs directly derived from adipose tissue (AT-EVs) are still limited. Here, we provide a systemic summary on the key components and functions of EVs derived from healthy adipose tissue, showing their significance on the tissue recovery and metabolic homeostasis regulation. Also, we discuss the harmful influences of EVs derived from obese adipose tissue on the distal organs. Furthermore, we elucidate the potential applications and constraints of EVs from healthy patients lipoaspirates as therapeutic agents, highlighting the potential of AT-EVs as a valuable biological material with broad prospects for future clinical use.
Collapse
Affiliation(s)
- Yue Han
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing, China
| | - Sheng Ye
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing, China
| | - Bowen Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Life Sciences, Westlake University, Hangzhou, China
| |
Collapse
|
5
|
Stojchevski R, Chandrasekaran P, Hadzi-Petrushev N, Mladenov M, Avtanski D. Adipose Tissue Dysfunction Related to Climate Change and Air Pollution: Understanding the Metabolic Consequences. Int J Mol Sci 2024; 25:7849. [PMID: 39063092 PMCID: PMC11277516 DOI: 10.3390/ijms25147849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity, a global pandemic, poses a major threat to healthcare systems worldwide. Adipose tissue, the energy-storing organ during excessive energy intake, functions as a thermoregulator, interacting with other tissues to regulate systemic metabolism. Specifically, brown adipose tissue (BAT) is positively associated with an increased resistance to obesity, due to its thermogenic function in the presence of uncoupled protein 1 (UCP1). Recently, studies on climate change and the influence of environmental pollutants on energy homeostasis and obesity have drawn increasing attention. The reciprocal relationship between increasing adiposity and increasing temperatures results in reduced adaptive thermogenesis, decreased physical activity, and increased carbon footprint production. In addition, the impact of climate change makes obese individuals more prone to developing type 2 diabetes mellitus (T2DM). An impaired response to heat stress, compromised vasodilation, and sweating increase the risk of diabetes-related comorbidities. This comprehensive review provides information about the effects of climate change on obesity and adipose tissue, the risk of T2DM development, and insights into the environmental pollutants causing adipose tissue dysfunction and obesity. The effects of altered dietary patterns on adiposity and adaptation strategies to mitigate the detrimental effects of climate change are also discussed.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10003, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | | | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.); (M.M.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.); (M.M.)
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10003, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
6
|
Kotańska M, Zadrożna M, Kubacka M, Mika K, Szczepańska K, Nowak B, Alesci A, Miller A, Lauriano ER, Kieć-Kononowicz K. The Effect of KSK-94, a Dual Histamine H 3 and Sigma-2 Receptor Ligand, on Adipose Tissue in a Rat Model of Developing Obesity. Pharmaceuticals (Basel) 2024; 17:858. [PMID: 39065709 PMCID: PMC11280160 DOI: 10.3390/ph17070858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/17/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Numerous studies highlight the critical role that neural histamine plays in feeding behavior, which is controlled by central histamine H3 and H1 receptors. This is the fundamental motivation for the increased interest in creating histamine H3 receptor antagonists as anti-obesity medications. On the other hand, multiple other neurotransmitter systems have been identified as pharmacotherapeutic targets for obesity, including sigma-2 receptor systems. Interestingly, in our previous studies in the rat excessive eating model, we demonstrated a significant reduction in the development of obesity using dual histamine H3/sigma-2 receptor ligands. Moreover, we showed that compound KSK-94 (structural analog of Abbott's A-331440) reduced the number of calories consumed, and thus acted as an anorectic compound. Therefore, in this study, we extended the previous research and studied the influence of KSK-94 on adipose tissue collected from animals from our previous experiment. METHODS Visceral adipose tissue was collected from four groups of rats (standard diet + vehicle, palatable diet + vehicle, palatable diet + KSK-94, and palatable diet + bupropion/naltrexone) and subjected to biochemical, histopathological, and immunohistochemical studies. RESULTS The obtained results clearly indicate that compound KSK-94 prevented the hypertrophy and inflammation of visceral adipose tissue, normalized the levels of leptin, resistin and saved the total reduction capacity of adipose tissue, being more effective than bupropion/naltrexon in these aspects. Moreover, KSK-94 may induce browning of visceral white adipose tissue. CONCLUSION Our study suggests that dual compounds with a receptor profile like KSK-94, i.e., targeting histamine H3 receptor and, to a lesser extent, sigma-2 receptor, could be attractive therapeutic options for patients at risk of developing obesity or with obesity and some metabolic disorders. However, more studies are required to determine its safety profile and the exact mechanism of action of KSK-94.
Collapse
Affiliation(s)
- Magdalena Kotańska
- Department of Pharmacological Screening, Jagiellonian University Medical College, Medyczna 9, PL 30-688 Kraków, Poland;
| | - Monika Zadrożna
- Department of Cytobiology, Jagiellonian University Medical College, Medyczna 9, PL 30-688 Kraków, Poland; (M.Z.); (B.N.)
| | - Monika Kubacka
- Department of Pharmacodynamics, Jagiellonian University Medical College, Medyczna 9, PL 30-688 Kraków, Poland;
| | - Kamil Mika
- Department of Pharmacological Screening, Jagiellonian University Medical College, Medyczna 9, PL 30-688 Kraków, Poland;
| | - Katarzyna Szczepańska
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343 Kraków, Poland;
| | - Barbara Nowak
- Department of Cytobiology, Jagiellonian University Medical College, Medyczna 9, PL 30-688 Kraków, Poland; (M.Z.); (B.N.)
| | - Alessio Alesci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, I-98166 Messina, Italy; (A.A.); (E.R.L.)
| | - Anthea Miller
- Department of Veterinary Sciences, University of Messina, I-98168 Messina, Italy;
| | - Eugenia Rita Lauriano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, I-98166 Messina, Italy; (A.A.); (E.R.L.)
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, PL 30-688 Kraków, Poland;
| |
Collapse
|
7
|
Kagdi S, Lyons SA, Beaudry JL. The interplay of glucose-dependent insulinotropic polypeptide in adipose tissue. J Endocrinol 2024; 261:e230361. [PMID: 38579777 PMCID: PMC11103678 DOI: 10.1530/joe-23-0361] [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: 11/17/2023] [Accepted: 04/03/2024] [Indexed: 04/07/2024]
Abstract
Adipose tissue was once known as a reservoir for energy storage but is now considered a crucial organ for hormone and energy flux with important effects on health and disease. Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone secreted from the small intestinal K cells, responsible for augmenting insulin release, and has gained attention for its independent and amicable effects with glucagon-like peptide 1 (GLP-1), another incretin hormone secreted from the small intestinal L cells. The GIP receptor (GIPR) is found in whole adipose tissue, whereas the GLP-1 receptor (GLP-1R) is not, and some studies suggest that GIPR action lowers body weight and plays a role in lipolysis, glucose/lipid uptake/disposal, adipose tissue blood flow, lipid oxidation, and free-fatty acid (FFA) re-esterification, which may or may not be influenced by other hormones such as insulin. This review summarizes the research on the effects of GIP in adipose tissue (distinct depots of white and brown) using cellular, rodent, and human models. In doing so, we explore the mechanisms of GIPR-based medications for treating metabolic disorders, such as type 2 diabetes and obesity, and how GIPR agonism and antagonism contribute to improvements in metabolic health outcomes, potentially through actions in adipose tissues.
Collapse
Affiliation(s)
- Samrin Kagdi
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sulayman A Lyons
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jacqueline L Beaudry
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Palacio PL, Greenwald J, Nguyen KT, Shantaram D, Butsch BL, Kim Y, Dattu MH, Noria S, Brethauer SA, Needleman BJ, Wysocki V, Hsueh W, Reátegui E, Magaña SM. Novel multiparametric bulk and single EV pipeline for adipose cell-specific biomarker discovery in paired human biospecimens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.18.590172. [PMID: 38659953 PMCID: PMC11042368 DOI: 10.1101/2024.04.18.590172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Obesity is a global health crisis that contributes to morbidity and mortality worldwide. Obesity's comorbid association with a variety of diseases, from metabolic syndrome to neurodegenerative disease, underscores the critical need to better understand the pathobiology of obesity. Adipose tissue, once seen as an inert storage depot, is now recognized as an active endocrine organ, regulating metabolic and systemic homeostasis. Recent studies spotlight the theranostic utility of extracellular vesicles (EVs) as novel biomarkers and drivers of disease, including obesity-related complications. Adipose-derived EVs (ADEVs) have garnered increased interest for their roles in diverse diseases, however robust isolation and characterization protocols for human, cell-specific EV subsets are limited. Herein, we directly address this technical challenge by establishing a multiparametric analysis framework that leverages bulk and single EV characterization, mRNA phenotyping and proteomics of human ADEVs directly from paired visceral adipose tissue, cultured mature adipocyte conditioned media, and plasma from obese subjects undergoing bariatric surgery. Importantly, rigorous EV phenotyping at the tissue and cell-specific level identified top 'adipose liquid biopsy' candidates that were validated in circulating plasma EVs from the same patient. In summary, our study paves the way toward a tissue and cell-specific, multiparametric framework for studying tissue and circulating adipose EVs in obesity-driven disease.
Collapse
|
9
|
Khandayataray P, Samal D, Murthy MK. Arsenic and adipose tissue: an unexplored pathway for toxicity and metabolic dysfunction. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:8291-8311. [PMID: 38165541 DOI: 10.1007/s11356-023-31683-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
Arsenic-contaminated drinking water can induce various disorders by disrupting lipid and glucose metabolism in adipose tissue, leading to insulin resistance. It inhibits adipocyte development and exacerbates insulin resistance, though the precise impact on lipid synthesis and lipolysis remains unclear. This review aims to explore the processes and pathways involved in adipogenesis and lipolysis within adipose tissue concerning arsenic-induced diabetes. Although arsenic exposure is linked to type 2 diabetes, the specific role of adipose tissue in its pathogenesis remains uncertain. The review delves into arsenic's effects on adipose tissue and related signaling pathways, such as SIRT3-FOXO3a, Ras-MAP-AP-1, PI(3)-K-Akt, endoplasmic reticulum stress proteins, CHOP10, and GPCR pathways, emphasizing the role of adipokines. This analysis relies on existing literature, striving to offer a comprehensive understanding of different adipokine categories contributing to arsenic-induced diabetes. The findings reveal that arsenic detrimentally impacts white adipose tissue (WAT) by reducing adipogenesis and promoting lipolysis. Epidemiological studies have hinted at a potential link between arsenic exposure and obesity development, with limited research suggesting a connection to lipodystrophy. Further investigations are needed to elucidate the mechanistic association between arsenic exposure and impaired adipose tissue function, ultimately leading to insulin resistance.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha, 752057, India
| | - Dibyaranjan Samal
- Department of Biotechnology, Sri Satya Sai University of Technical and Medical Sciences, Sehore, Madhya Pradesh, 466001, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab, 140401, India.
| |
Collapse
|
10
|
Abbasi K, Zarezadeh R, Valizadeh A, Mehdizadeh A, Hamishehkar H, Nouri M, Darabi M. White-brown adipose tissue interplay in polycystic ovary syndrome: Therapeutic avenues. Biochem Pharmacol 2024; 220:116012. [PMID: 38159686 DOI: 10.1016/j.bcp.2023.116012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
This study highlights the therapeutic potential of activating brown adipose tissue (BAT) for managing polycystic ovary syndrome (PCOS), a prevalent endocrine disorder associated with metabolic and reproductive abnormalities. BAT plays a crucial role in regulating energy expenditure and systemic insulin sensitivity, making it an attractive target for the treatment of obesity and metabolic diseases. Recent research suggests that impaired BAT function and mass may contribute to the link between metabolic disturbances and reproductive issues in PCOS. Additionally, abnormal white adipose tissue (WAT) can exacerbate these conditions by releasing adipokines and nonesterified fatty acids. In this review, we explored the impact of WAT changes on BAT function in PCOS and discussed the potential of BAT activation as a therapeutic strategy to improve PCOS symptoms. We propose that BAT activation holds promise for managing PCOS; however, further research is needed to confirm its efficacy and to develop clinically feasible methods for BAT activation.
Collapse
Affiliation(s)
- Khadijeh Abbasi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Valizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, Germany.
| |
Collapse
|
11
|
Engin A. Adipose Tissue Hypoxia in Obesity: Clinical Reappraisal of Hypoxia Hypothesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:329-356. [PMID: 39287857 DOI: 10.1007/978-3-031-63657-8_11] [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: 09/19/2024]
Abstract
Obese subjects exhibit lower adipose tissue oxygen consumption in accordance with the lower adipose tissue blood flow. Thereby, compared to lean subjects, obese individuals have almost half lower capillary density and more than half lower vascular endothelial growth factor (VEGF). The VEGF expression together with hypoxia-inducible transcription factor-1 alpha (HIF-1α) activity also requires phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR)-mediated signaling. Especially HIF-1α is an important signaling molecule for hypoxia to induce the inflammatory responses. Hypoxia contributes to several biological functions, such as angiogenesis, cell proliferation, apoptosis, inflammation, and insulin resistance (IR). Pathogenesis of obesity-related comorbidities is attributed to intermittent hypoxia (IH), which is mostly observed in visceral obesity. Proinflammatory phenotype of the adipose tissue is a crucial link between IH and the development of IR. Inhibition of adaptive unfolded protein response (UPR) in hypoxia increases β cell death. Moreover, deletion of HIF-1α worsens β cell function. Oxidative stress, as well as the release of proinflammatory cytokines/adipokines in obesity, is proportional to the severity of IH. Reactive oxygen species (ROS) generation at mitochondria is responsible for propagation of the hypoxic signal; however, mitochondrial ROS production is required for hypoxic HIF-1α protein stabilization. Alterations in oxygen availability of adipose tissue directly affect the macrophage polarization and are responsible for the dysregulated adipocytokines production in obesity. Hypoxia both inhibits adipocyte differentiation from preadipocytes and macrophage migration from the hypoxic adipose tissue. Upon reaching a hypertrophic threshold beyond the adipocyte fat loading capacity, excess extracellular matrix (ECM) components are deposited, causing fibrosis. HIF-1α initiates the whole pathological process of fibrosis and inflammation in the obese adipose tissue. In addition to stressed adipocytes, hypoxia contributes to immune cell migration and activation which further aggravates adipose tissue fibrosis. Therefore, targeting HIF-1α might be an efficient way to suppress hypoxia-induced pathological changes in the ECM. The fibrosis score of adipose tissue correlates negatively with the body mass index and metabolic parameters. Inducers of browning/beiging adipocytes and adipokines, as well as modulations of matrix remodeling enzyme inhibitors, and associated gene regulators, are potential pharmacological targets for treating obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
12
|
Vijverberg SJ, Kampouras A, Nayir Büyükşahin H, Makrinioti H, Petrarca L, Schmidt M, Schreck LD, Urbantat RM, Beydon N, Goutaki M, Lavizzari A, Proesmans M, Schramm D, Stahl M, Zacharasiewicz A, Moeller A, Pijnenburg MW. ERS International Congress 2023: highlights from the Paediatrics Assembly. ERJ Open Res 2024; 10:00853-2023. [PMID: 38410713 PMCID: PMC10895434 DOI: 10.1183/23120541.00853-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 02/28/2024] Open
Abstract
Respiratory health in children is essential for general wellbeing and healthy development in the short and long term. It is well known that many respiratory diseases in adulthood have their origins in early life, and therefore research on prevention of respiratory diseases and management of children with respiratory diseases will benefit patients during the full life course. Scientific and clinical advances in the field of respiratory health are moving at a fast pace. This article summarises some of the highlights in paediatric respiratory medicine presented at the hybrid European Respiratory Society (ERS) International Congress 2023 which took place in Milan (Italy). Selected sessions are summarised by Early Career Members of the Paediatrics Assembly (Assembly 7) under the supervision of senior ERS officers, and cover a wide range of research areas in children, including respiratory physiology and sleep, asthma and allergy, cystic fibrosis, respiratory infection and immunology, neonatology and intensive care, respiratory epidemiology and bronchology.
Collapse
Affiliation(s)
- Susanne J.H. Vijverberg
- Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Pediatric Pulmonology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Asterios Kampouras
- Paediatric Pulmonology Department, 424 General Military Hospital, Thessaloniki, Greece
| | - Halime Nayir Büyükşahin
- Division of Pulmonology, Department of Paediatrics, Mardin Training and Research Hospital, Mardin, Turkey
| | - Heidi Makrinioti
- Department of Emergency Medicine, Harvard Medical School, Boston, MA, USA
| | - Laura Petrarca
- Translational and Precision Medicine Department, “Sapienza” University of Rome, Rome, Italy
- Maternal Infantile and Urological Sciences Department, “Sapienza” University of Rome, Rome, Italy
| | - Mehtap Schmidt
- Department of Pediatrics, Teaching Hospital of the University of Vienna, Wilhelminen Hospital, Vienna, Austria
| | - Leonie D. Schreck
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Ruth M. Urbantat
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Nicole Beydon
- Assistance Publique-Hôpitaux de Paris, Unité Fonctionnelle de Physiologie – Explorations Fonctionnelles Respiratoires et du Sommeil, Hôpital Armand Trousseau, Paris, France
- INSERM, U 938, Centre de Recherche Saint Antoine, Hôpital Saint-Antoine, Paris, France
| | - Myrofora Goutaki
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Anna Lavizzari
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marijke Proesmans
- Division of Woman and Child, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Dirk Schramm
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Mirjam Stahl
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Angela Zacharasiewicz
- Department of Pediatrics, Teaching Hospital of the University of Vienna, Wilhelminen Hospital, Vienna, Austria
| | - Alexander Moeller
- Department of Paediatric Pulmonology, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marielle W. Pijnenburg
- Department of Paediatrics, Division of Paediatric Respiratory Medicine and Allergology, Erasmus MC – Sophia Children's Hospital, University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
13
|
Prajapati S. Advances in the Management of Diabetes and Overweight using Incretin-based Pharmacotherapies. Curr Diabetes Rev 2024; 20:e131123223544. [PMID: 37962047 DOI: 10.2174/0115733998256797231009062744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/20/2023] [Accepted: 08/30/2023] [Indexed: 11/15/2023]
Abstract
Throughout the previous three decades, the secretion of glucagon-like peptide-1 hormone has attracted much attention to attain possible therapy goals for the treatment of both hypoglycaemic along type II diabetes militates and overweight. The pharmaceutical generation of peptides similar to hypoglycaemia-based medicines is exemplified by agonists of the GLP- 1R (Glucagon-like peptide-1 receptors). Pharmacokinetic profiles are continuously being improved, beginning with the native hormone with a two- to three-minute quarter and progressing through growth every day with once-drug combinations. Due to contradictory data that indicate stimulation or inhibition of the Glucagon-like peptide receptor, the Glucose-dependent insulin tropic peptide receptor offers favorable effects on systemic metabolism. The recent Glp-1R (Glucagon-like peptide-1 receptor-) targeting monomolecular drugs has demonstrated therapeutic effectiveness and has stoked interest in Glucose-dependent insulin tropic polypeptide antagonism as a treatment for overweight and diabetes mellitus. These drugs have been shown to dramatically improve carbohydrates with body weight management in sick people who have obesity and type II diabetes mellitus. In this study, recent breakthroughs in compelling therapeutic interventions are discussed, and the biology and pharmacology of the glucose-like peptide are reviewed.
Collapse
Affiliation(s)
- Shatrudhan Prajapati
- Department of Pharmacy, Golgotias University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
14
|
Geiger K, Muendlein A, Leiherer A, Gaenger S, Brandtner EM, Wabitsch M, Fraunberger P, Drexel H, Heinzle C. Myricetin attenuates hypoxia-induced inflammation in human adipocytes. Mol Biol Rep 2023; 50:9833-9843. [PMID: 37843712 DOI: 10.1007/s11033-023-08865-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/27/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Adipose tissue hypoxia plays a crucial role in the development of chronic low-grade systemic inflammation which has been associated with the pathogenesis of obesity-related diseases. Myricetin is a natural compound present in numerous plant-based foods with presumed anti-inflammatory and beneficial health effects. The impact of this flavonoid on hypoxia-induced expression of inflammatory adipokines and hypoxia-regulated pathways is unknown so far and has been addressed in the present study. METHODS Differentiated human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes were cultured with or without myricetin under normoxic and hypoxic conditions for varying time periods. The effect of hypoxia and myricetin on the expression of the investigated adipokines was measured by real-time RT-PCR. Western blot analysis was used for the detection of transcription factors involved in hypoxia-regulated pathways. RESULTS Myricetin interfered in the hypoxia-induced regulation of adipokines and the underlying pathways, which are involved in transmitting the inflammatory response. It strongly repressed hypoxia-induced expression of apelin, leptin, chemerin, asprosin, and DPP-4 and HIF-1α accumulation in the nucleus was diminished. Furthermore, the activation of the key regulators in the inflammatory response NF-κB, Akt, and CREB was suppressed by myricetin under hypoxic conditions. Myricetin also decreased hypoxia-induced accumulation of the pro-tumorigenic transcription factors Snail and Slug in the nucleus. CONCLUSION Taken together, our results indicated that myricetin regulated hypoxia-induced expression of adipokines and hypoxia-regulated pathways in human adipocytes. Our study therefore provided evidence of the anti-inflammatory effects of myricetin in hypoxia-treated human adipocytes.
Collapse
Affiliation(s)
- Kathrin Geiger
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.
- Medical Central Laboratories, Feldkirch, Austria.
| | - Axel Muendlein
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | - Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Medical Central Laboratories, Feldkirch, Austria
- Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Stella Gaenger
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | - Eva Maria Brandtner
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | | | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
- Vorarlberger Landeskrankenhausbetriebsgesellschaft, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Christine Heinzle
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Medical Central Laboratories, Feldkirch, Austria
| |
Collapse
|
15
|
Kang GS, Jo HJ, Lee YR, Oh T, Park HJ, Ahn GO. Sensing the oxygen and temperature in the adipose tissues - who's sensing what? Exp Mol Med 2023; 55:2300-2307. [PMID: 37907745 PMCID: PMC10689767 DOI: 10.1038/s12276-023-01113-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 11/02/2023] Open
Abstract
Adipose tissues, composed of various cell types, including adipocytes, endothelial cells, neurons, and immune cells, are organs that are exposed to dynamic environmental challenges. During diet-induced obesity, white adipose tissues experience hypoxia due to adipocyte hypertrophy and dysfunctional vasculature. Under these conditions, cells in white adipose tissues activate hypoxia-inducible factor (HIF), a transcription factor that activates signaling pathways involved in metabolism, angiogenesis, and survival/apoptosis to adapt to such an environment. Exposure to cold or activation of the β-adrenergic receptor (through catecholamines or chemicals) leads to heat generation, mainly in brown adipose tissues through activating uncoupling protein 1 (UCP1), a proton uncoupler in the inner membrane of the mitochondria. White adipose tissues can undergo a similar process under this condition, a phenomenon known as 'browning' of white adipose tissues or 'beige adipocytes'. While UCP1 expression has largely been confined to adipocytes, HIF can be expressed in many types of cells. To dissect the role of HIF in specific types of cells during diet-induced obesity, researchers have generated tissue-specific knockout (KO) mice targeting HIF pathways, and many studies have commonly revealed that intact HIF-1 signaling in adipocytes and adipose tissue macrophages exacerbates tissue inflammation and insulin resistance. In this review, we highlight some of the key findings obtained from these transgenic mice, including Ucp1 KO mice and other models targeting the HIF pathway in adipocytes, macrophages, or endothelial cells, to decipher their roles in diet-induced obesity.
Collapse
Affiliation(s)
- Gi-Sue Kang
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Hye-Ju Jo
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Ye-Rim Lee
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Taerim Oh
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Hye-Joon Park
- College of Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - G-One Ahn
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea.
- College of Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea.
| |
Collapse
|
16
|
Osorio-Conles Ó, Jiménez A, Ibarzabal A, Balibrea JM, de Hollanda A, Vidal J. Limited Bariatric Surgery-induced Weight Loss in Subjects With Type 2 Diabetes: Predictor Variables in Adipose Tissue. J Clin Endocrinol Metab 2023; 108:e1205-e1213. [PMID: 37249080 DOI: 10.1210/clinem/dgad271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Indexed: 05/31/2023]
Abstract
CONTEXT The impact of type 2 diabetes mellitus (T2D) at baseline on limited weight loss (WL) after bariatric surgery (BS) remains controversial, and the potential underlying mechanisms incompletely understood. OBJECTIVE We aimed at gaining further insight on this relationship and identifying novel associations between adipose tissue (AT) parameters and short-term WL outcomes in subjects with or without T2D undergoing BS. METHODS Mid-term WL trajectories after BS have been evaluated in a cohort of 1659 subjects (cohort 1) with (n = 543) and without T2D (n = 1116). Paired subcutaneous and visceral AT samples were obtained from a cohort of 48 pairs of subjects with and without T2D matched for age, sex, BMI, and type of BS (cohort 2). Differences in AT parameters between groups were evaluated and potential associations with WL response explored. RESULTS T2D was independently associated with a 5% lesser mid-term WL in cohort 1, while HbA1c, insulin treatment, and number of T2D medications prior to BS were only related to short-term WL outcomes. In cohort 2, a number of differentially expressed genes in AT were identified between groups, while fat cell size and fibrosis were comparable. Subcutaneous ATG7 expression was found as an independent predictor of limited WL 1 year after surgery (β: -12.21 ± 4.41, P = .008) and its addition to a clinical model significantly improved the amount of WL variability explained (R2 = 0.131 vs R2 = 0.248, F change P = .009). CONCLUSION Our results highlight the importance of T2D as determinant of limited WL following BS and suggest that dysregulated macroautophagy in subcutaneous AT may contribute to this association.
Collapse
Affiliation(s)
- Óscar Osorio-Conles
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Amanda Jiménez
- Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Fisiopatologia de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Ainitze Ibarzabal
- Gastrointestinal Surgery Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - José María Balibrea
- Gastrointestinal Surgery Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Ana de Hollanda
- Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Fisiopatologia de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Josep Vidal
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
17
|
Lempesis IG, Varrias D, Sagris M, Attaran RR, Altin ES, Bakoyiannis C, Palaiodimos L, Dalamaga M, Kokkinidis DG. Obesity and Peripheral Artery Disease: Current Evidence and Controversies. Curr Obes Rep 2023; 12:264-279. [PMID: 37243875 PMCID: PMC10220347 DOI: 10.1007/s13679-023-00510-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 05/29/2023]
Abstract
PURPOSE OF REVIEW Obesity is a significant public health problem and a major risk factor for the development and progression of atherosclerosis and its cardiovascular manifestations. Lower extremity peripheral artery disease (PAD) affects 3%-10% of the Western population and, if left untreated, can lead to devastating outcomes with both an increased risk of morbidity and mortality. Interestingly, the association between obesity and PAD remains debatable. Whereas it is well known that PAD and obesity frequently overlap in the same patients, many studies have demonstrated a negative association between obesity and PAD and a protective effect of obesity on disease development and progression, a phenomenon described as the "obesity paradox." Possible mechanisms for this paradox may include genetic background, as assessed by mendelian randomization studies, adipose tissue dysfunction, and body fat distribution rather than adiposity, while other factors, such as sex, ethnicity, sarcopenia in the elderly population, or aggressive treatment of co-existing metabolic conditions in individuals with obesity compared to those with normal weight, could have some impact as well. RECENT RINDINGS Few reviews and meta-analyses examining systematically the relationship between obesity and PAD exist. The impact of PAD development due to the presence of obesity remains largely controversial. However, the most current evidence, backed by a recent meta-analysis, suggests a potential protective role of a higher body mass index on PAD-related complications and mortality. In this review, we discuss the association between obesity and PAD development, progression, and management, and the potential pathophysiologic mechanisms linking the two diseases.
Collapse
Affiliation(s)
- Ioannis G Lempesis
- Department of Biologic Chemistry, School of Medicine, National and Kapodistrian, University of Athens, Mikras Asias 75, 115 27, Athens, Greece.
| | - Dimitrios Varrias
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marios Sagris
- General Hospital of Nikaia, Piraeus, 184 54, Athens, Greece
| | - Robert R Attaran
- Section of Cardiovascular Medicine, Yale University/Yale New Haven Hospital, 06519, New Haven, CT, USA
| | - Elissa S Altin
- Section of Cardiovascular Medicine, Yale University/Yale New Haven Hospital, 06519, New Haven, CT, USA
| | - Christos Bakoyiannis
- Department of Surgery, Division of Vascular Surgery, Laikon General Hospital, National Kapodistrian University of Athens, 15772, Athens, Greece
| | - Leonidas Palaiodimos
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Dalamaga
- Department of Biologic Chemistry, School of Medicine, National and Kapodistrian, University of Athens, Mikras Asias 75, 115 27, Athens, Greece
| | - Damianos G Kokkinidis
- Section of Cardiovascular Medicine, Yale University/Yale New Haven Hospital, 06519, New Haven, CT, USA
| |
Collapse
|
18
|
Bubnovskaya L, Ganusevich I, Merentsev S, Osinsky D. CANCER-ASSOCIATED ADIPOCYTES AND PROGNOSTIC VALUE OF PREOPERATIVE NEUTROPHIL-LYMPHOCYTE RATIO IN GASTRIC CANCER. Exp Oncol 2023; 45:88-98. [PMID: 37417278 DOI: 10.15407/exp-oncology.2023.01.088] [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: 06/26/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND The neutrophil-to-lymphocyte ratio (NLR) turned out to be a routinely available marker capable to reflect the systemic inflammatory response created by a tumor. Gastric cancer (GC) grows in the anatomical vicinity of adipose tissue, which is also associated with low-grade inflammation. AIM To investigate the usefulness of the combined use of preoperative NLR and density of intratumoral cancer-associated adipocytes (CAAs) for predicting the disease outcome in GC patients. MATERIALS AND METHODS A total of 151 patients with GC were eligible for retrospective analysis between 2009 and 2015.NLR preoperative values were calculated. Perilipin expression in tumor tissue was examined immunohistochemically. RESULTS Low preoperative NLR is the most reliable prognostic factor for the favorable outcome for patients with low density of intratumoral CAAs. Patients with a high density of CCAs are at high risk of lethal outcomes independently of the value of preoperative NLR. CONCLUSION The results have clearly shown an association between preoperative NLR and the density of CAAs in the primary tumor of GC patients. The prognostic value of NLR is essentially modified by means of the individual density of intratumoral CAAs in GC patients.The elevated NLR could be of significant predictive potential for a negative prognosis for patients with tumors characterized by the high density of CAAs independently of BMI.
Collapse
Affiliation(s)
- L Bubnovskaya
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv 03022, Ukraine
| | - I Ganusevich
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv 03022, Ukraine
| | - S Merentsev
- City Clinical Oncological Center, Kyiv 03115, Ukraine
| | - D Osinsky
- City Clinical Oncological Center, Kyiv 03115, Ukraine
| |
Collapse
|
19
|
Lempesis IG, Hoebers N, Essers Y, Jocken JWE, Dineen R, Blaak EE, Manolopoulos KN, Goossens GH. Distinct inflammatory signatures of upper and lower body adipose tissue and adipocytes in women with normal weight or obesity. Front Endocrinol (Lausanne) 2023; 14:1205799. [PMID: 37455922 PMCID: PMC10338223 DOI: 10.3389/fendo.2023.1205799] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/02/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Upper and lower body fat accumulation poses an opposing obesity-related cardiometabolic disease risk. Depot-differences in subcutaneous adipose tissue (SAT) function may underlie these associations. We aimed to investigate the inflammatory signatures of abdominal (ABD) and femoral (FEM) SAT in postmenopausal women with normal weight or obesity. Methods We included 23 postmenopausal women with normal weight (n = 13) or obesity (n = 10). In vivo secretion of adipokines from ABD and FEM SAT was measured using the arterio-venous balance technique. Adipokine gene expression and adipocyte morphology were examined in ABD and FEM SAT. Furthermore, adipokine expression and secretion were investigated in vitro using differentiated human primary ABD and FEM subcutaneous adipocytes derived from the study participants. Results Plasma leptin and plasminogen activator inhibitor (PAI)-1 concentrations were higher, and ABD and FEM adipocytes were larger in women with obesity than normal weight. No differences in adipocyte size and blood flow were apparent between ABD and FEM SAT. We found significant release of leptin and monocyte chemoattractant protein (MCP)-1 from ABD and FEM SAT, with higher fractional release of MCP-1 from ABD than FEM SAT. Gene expression of leptin, PAI-1, and tumor necrosis factor-α was lower in ABD than FEM SAT and higher in women with obesity than normal weight. In ABD adipocytes, interleukin-6, PAI-1, and leptin gene expression were higher, while adiponectin and dipeptidyl-peptidase-4 gene expression were lower than in FEM adipocytes. Finally, ABD adipocytes secreted less MCP-1 compared to FEM adipocytes. Discussion These findings demonstrate that upper and lower body SAT and adipocytes are characterized by distinct inflammatory signatures in postmenopausal women, which seem independent of adipocyte size.
Collapse
Affiliation(s)
- Ioannis G. Lempesis
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Nicole Hoebers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
| | - Yvonne Essers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
| | - Johan W. E. Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
| | - Rosemary Dineen
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Ellen E. Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
| | - Konstantinos N. Manolopoulos
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
| |
Collapse
|
20
|
Lempesis IG, Georgakopoulou VE. Physiopathological mechanisms related to inflammation in obesity and type 2 diabetes mellitus. World J Exp Med 2023; 13:7-16. [PMID: 37396883 PMCID: PMC10308320 DOI: 10.5493/wjem.v13.i3.7] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/15/2023] [Accepted: 04/10/2023] [Indexed: 06/16/2023] Open
Abstract
Overweight, obesity, and type 2 diabetes mellitus pose global health problems that are ever-increasing. A chronic low-grade inflammatory status and the presence of various pro-inflammatory markers either in circulation or within dysfunctional metabolic tissues are well established. The presence of these factors can, to some extent, predict disease development and progression. A central role is played by the presence of dysfunctional adipose tissue, liver dysfunction, and skeletal muscle dysfunction, which collectively contribute to the increased circulatory levels of proinflammatory factors. Weight loss and classical metabolic interventions achieve a decrease in many of these factors' circulating levels, implying that a better understanding of the processes or even the modulation of inflammation may alleviate these diseases. This review suggests that inflammation plays a significant role in the development and progression of these conditions and that measuring inflammatory markers may be useful for assessing disease risk and development of future treatment methods.
Collapse
Affiliation(s)
- Ioannis G Lempesis
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Athens 11527, Greece
| | | |
Collapse
|
21
|
Zhou C, Huang YQ, Da MX, Jin WL, Zhou FH. Adipocyte-derived extracellular vesicles: bridging the communications between obesity and tumor microenvironment. Discov Oncol 2023; 14:92. [PMID: 37289328 PMCID: PMC10250291 DOI: 10.1007/s12672-023-00704-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/26/2023] [Indexed: 06/09/2023] Open
Abstract
By the year 2035 more than 4 billion people might be affected by obesity and being overweight. Adipocyte-derived Extracellular Vesicles (ADEVs/ADEV-singular) are essential for communication between the tumor microenvironment (TME) and obesity, emerging as a prominent mechanism of tumor progression. Adipose tissue (AT) becomes hypertrophic and hyperplastic in an obese state resulting in insulin resistance in the body. This modifies the energy supply to tumor cells and simultaneously stimulates the production of pro-inflammatory adipokines. In addition, obese AT has a dysregulated cargo content of discharged ADEVs, leading to elevated amounts of pro-inflammatory proteins, fatty acids, and carcinogenic microRNAs. ADEVs are strongly associated with hallmarks of cancer (proliferation and resistance to cell death, angiogenesis, invasion, metastasis, immunological response) and may be useful as biomarkers and antitumor therapy strategy. Given the present developments in obesity and cancer-related research, we conclude by outlining significant challenges and significant advances that must be addressed expeditiously to promote ADEVs research and clinical applications.
Collapse
Affiliation(s)
- Chuan Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000 People’s Republic of China
| | - Yu-Qian Huang
- Department of Center of Medical Cosmetology, Chengdu Second People’s Hospital, Chengdu, 610017 People’s Republic of China
| | - Ming-Xu Da
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000 People’s Republic of China
| | - Wei-Lin Jin
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Feng-Hai Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Department of Urology, Gansu Provincial Hospital, Lanzhou, 730000 People’s Republic of China
| |
Collapse
|
22
|
Lempesis IG, Georgakopoulou VE. Implications of obesity and adiposopathy on respiratory infections; focus on emerging challenges. World J Clin Cases 2023; 11:2925-2933. [PMID: 37215426 PMCID: PMC10198078 DOI: 10.12998/wjcc.v11.i13.2925] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 04/25/2023] Open
Abstract
Obesity is characterized by excessive adipose tissue accumulation, which impacts physiological, metabolic, and immune functions. Several respiratory infections, including bacterial pneumonia, influenza, and coronavirus disease 2019, appear to be linked to unfavorable results in individuals with obesity. These may be attributed to the direct mechanical/physiological effects of excess body fat on the lungs’ function. Notably, adipose tissue dysfunction is associated with a low-grade chronic inflammatory status and hyperleptinemia, among other characteristics. These have all been linked to immune system dysfunction and weakened immune responses to these infections. A better understanding and clinical awareness of these risk factors are necessary for better disease outcomes.
Collapse
Affiliation(s)
- Ioannis G Lempesis
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Athens 11527, Greece
| | | |
Collapse
|
23
|
Clemente-Suárez VJ, Redondo-Flórez L, Beltrán-Velasco AI, Martín-Rodríguez A, Martínez-Guardado I, Navarro-Jiménez E, Laborde-Cárdenas CC, Tornero-Aguilera JF. The Role of Adipokines in Health and Disease. Biomedicines 2023; 11:biomedicines11051290. [PMID: 37238961 DOI: 10.3390/biomedicines11051290] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Adipokines are cell-signaling proteins secreted by adipose tissue that has been related to a low-grade state of inflammation and different pathologies. The present review aims to analyze the role of adipokines in health and disease in order to understand the important functions and effects of these cytokines. For this aim, the present review delves into the type of adipocytes and the cytokines produced, as well as their functions; the relations of adipokines in inflammation and different diseases such as cardiovascular, atherosclerosis, mental diseases, metabolic disorders, cancer, and eating behaviors; and finally, the role of microbiota, nutrition, and physical activity in adipokines is discussed. This information would allow for a better understanding of these important cytokines and their effects on body organisms.
Collapse
Affiliation(s)
| | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, C/Tajo s/n, 28670 Madrid, Spain
| | - Ana Isabel Beltrán-Velasco
- Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248 Madrid, Spain
| | | | - Ismael Martínez-Guardado
- BRABE Group, Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248 Madrid, Spain
| | | | | | | |
Collapse
|
24
|
Namdev G, Choudhari R, Khan AA, Ali N, Rashid S, Singh HO. Impact of inflammatory cytokine and adipokine gene variations in the development of HIV-associated lipodystrophy. J Gene Med 2023:e3512. [PMID: 37186064 DOI: 10.1002/jgm.3512] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/23/2023] [Accepted: 03/31/2023] [Indexed: 05/17/2023] Open
Abstract
Cytokines affect lipid and glucose metabolism and also alter the body's habitus. They play a role in the development of lipodystrophy syndrome. Adipocytes secrete the pro-inflammatory cytokines IL-1, TNF-α and IL-6. The plasma cytokine concentration is associated with the percentage and distribution of fat tissue in the body. The metabolic disturbances are strongly associated with increased levels of pro-inflammatory cytokines (IL-1, IL-6 and TNF-α). Plasma levels of cytokines such as TNF-α, IL-6 and leptin were found to be increased while plasma resistin levels were found to be variable in patients suffering from obesity and type II diabetes mellitus. Until now, limited information has been available on the polymorphism of cytokine and adipokine genes in patients of HIV-associated lipodystrophy (HIVLD), which can contribute to individual variations in susceptibility to metabolic diseases, especially to HIVLD. Hence, we studied the association of cytokine and adipokine gene polymorphisms in various diseases and their impact on HIVLD. We carry out an extensive search using several databases, including PubMed, EMBASE and Google Scholar. The distribution of cytokine and adipokine gene polymorphisms and their expression levels varied among various populations. We examined the variants of cytokine and adipokine genes, which can contribute to individual variations in susceptibility to metabolic diseases, especially to HIVLD. In the current review, we present a brief account of the risk factors of HIVLD, the pathogenesis of HIVLD and the polymorphism of cytokine and adipokine genes in various diseases with special reference to their impact on HIVLD.
Collapse
Affiliation(s)
- Goldi Namdev
- Division of Molecular Biology, National AIDS Research Institute, Pune, India
| | - Ranjana Choudhari
- Division of Molecular Biology, National AIDS Research Institute, Pune, India
| | - Abdul Arif Khan
- Division of Microbiology, National AIDS Research Institute, Pune, India
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Summya Rashid
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Hari Om Singh
- Division of Molecular Biology, National AIDS Research Institute, Pune, India
| |
Collapse
|
25
|
Lempesis IG, Karlafti E, Papalexis P, Fotakopoulos G, Tarantinos K, Lekakis V, Papadakos SP, Cholongitas E, Georgakopoulou VE. COVID-19 and liver injury in individuals with obesity. World J Gastroenterol 2023; 29:908-916. [PMID: 36844135 PMCID: PMC9950870 DOI: 10.3748/wjg.v29.i6.908] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/18/2022] [Accepted: 01/09/2023] [Indexed: 02/10/2023] Open
Abstract
Coronavirus disease 2019 is an infectious disease caused by the severe acute respiratory syndrome coronavirus 2 that manifests as a variety of clinical manifestations, including liver damage commonly detected by a hepatocellular pattern from liver function tests. Liver injury is associated with a worse prognosis overall. Conditions associated with the severity of the disease include obesity and cardiometabolic comorbidities, which are also associated with nonalcoholic fatty liver disease (NAFLD). The presence of NAFLD, similarly to obesity, is associated with an unfavourable impact on the coronavirus disease 2019 outcome. Individuals with these conditions could present with liver damage and elevated liver function tests due to direct viral cytotoxicity, systemic inflammation, ischemic or hypoxic liver damage or drug side effects. However, liver damage in the setting of NAFLD could also be attributed to a pre-existing chronic low-grade inflammation associated with surplus and dysfunctional adipose tissue in these individuals. Here we investigate the hypothesis that a pre-existing inflammatory status is exacerbated after severe acute respiratory syndrome coronavirus 2 infection, which embodies a second hit to the underestimated liver damage.
Collapse
Affiliation(s)
- Ioannis G Lempesis
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, United Kingdom
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht 616 6200, Netherlands
| | - Eleni Karlafti
- Department of Emergency, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki 546 21, Greece
| | - Petros Papalexis
- Unit of Endocrinology, First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
- Department of Biomedical Sciences, University of West Attica, Athens 12243, Greece
| | - George Fotakopoulos
- Department of Neurosurgery, General University Hospital of Larisa, Larisa 41221, Greece
| | | | - Vasileios Lekakis
- Department of Gastroenterology, Laiko General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Stavros P Papadakos
- Department of Gastroenterology, Laiko General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Evangelos Cholongitas
- First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | | |
Collapse
|
26
|
Huang B, Wan Q, Li T, Yu L, Du W, Calhoun C, Leong KW, Qiang L. Polycationic PAMAM ameliorates obesity-associated chronic inflammation and focal adiposity. Biomaterials 2023; 293:121850. [PMID: 36450630 DOI: 10.1016/j.biomaterials.2022.121850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/29/2022]
Abstract
As a surging public health crisis, obesity and overweight predispose individuals to various severe comorbidities contributed by the accompanying chronic inflammation. However, few options exist for tackling chronic inflammation in obesity or inhibiting depot-specific adiposity. Here, we report that polycationic polyamidoamine (PAMAM) treatment can improve both aspects of obesity. With the discovery that the plasma cell-free RNA (cfRNA) level is elevated in obese subjects, we applied the cationic PAMAM generation 3 (P-G3) scavenger to treat diet-induced obese (DIO) mice. Intraperitoneal delivery of P-G3 alleviated the chronic inflammation in DIO mice and reduced their body weight, resulting in improved metabolic functions. To further enhance the applicability of P-G3, we complexed P-G3 with human serum albumin (HSA) to attain a sustained release, which showed consistent benefits in treating DIO mice. Local injection of HSA-PG3 into subcutaneous fat completely restricted the distribution of the complex within the targeted depot and reduced focal adiposity. Our study illuminates a promising cationic strategy to ameliorate chronic inflammation in obesity and target local adiposity.
Collapse
Affiliation(s)
- Baoding Huang
- Department of Orthopaedic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510000, China; Department of Biomedical Engineering, Columbia University, New York, NY, 10032, USA
| | - Qianfen Wan
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Tianyu Li
- Department of Biomedical Engineering, Columbia University, New York, NY, 10032, USA
| | - Lexiang Yu
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Wen Du
- Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Carmen Calhoun
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10032, USA.
| | - Li Qiang
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
27
|
Ali S, Alam R, Ahmad MK, Ahmad M, Ahsan H, Khan MM, Khan S. Evaluation of serum adipokines (omentin-1 and visfatin) in coronary artery disease at a North Indian hospital. Endocr Regul 2023; 57:262-268. [PMID: 38127689 DOI: 10.2478/enr-2023-0029] [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] [Indexed: 12/23/2023] Open
Abstract
Objective. Adipose tissue is considered to be an endocrine organ that secretes bioactive substances known as adipokines that contribute to the pathophysiology of metabolic and coronary diseases related to obesity. In this study, various novel biomarkers, such as inflammatory markers that are pro-inflammatory (visfatin) and anti-inflammatory (omentin-1), as prognostic indicators for people with coronary artery disease (CAD) were investigated. Methods. In this study, 30 diabetic patients with CAD, 30 diabetic patients without CAD, and 30 healthy control counterparts were included. Serum omentin and visfatin concentrations were evaluated by solid-phase enzyme linked immunosorbent assay (ELISA) kit. Patients with established diagnosis of CAD based on angiography, ECG, and elevated cardiac marker level were included into the study. Patients with cardioembolic stroke, cerebral venous sinus thrombosis, CNS vasculitis, and hemorrhage due to trauma, tumor, vascular malformation, and coagulopathy were excluded. Results. The serum omentin-1 levels were significantly higher in the healthy controls in comparison with the diabetic group (p<0.0001) and serum visfatin levels were significantly higher in the diabetic group in comparison with the healthy controls (p<0.0001). The serum omentin levels were significantly higher in the diabetic group in comparison with the cardio-diabetic group (p<0.0001) and serum visfatin levels were significantly higher in the cardio-diabetic group in comparison with the diabetic group (p<0.0001). The serum omentin-1 showed negative correlation with the serum visfatin in the cardio-diabetic group. Conclusion. The adipokines, such as omentin and visfatin, may be good therapeutic candidates in preventing or ameliorating CAD.
Collapse
Affiliation(s)
- Saif Ali
- 1Department of Biochemistry, Integral Institute of Medical Science and Research, Integral University, Lucknow, India
| | - Roshan Alam
- 1Department of Biochemistry, Integral Institute of Medical Science and Research, Integral University, Lucknow, India
| | | | - Mukhtar Ahmad
- 3Department of Medicine, Integral Institute of Medical Science and Research, Integral University, Lucknow, India
| | - Haseeb Ahsan
- 4Department of Biochemistry, Faculty of Dentistry, Jamia Millia Islamia, New Delhi, India
| | - Mohammad Mustafa Khan
- 5Department of Basic Medical Sciences, Integral Institute of Allied Health Sciences and Research, Integral University, Lucknow, India
| | - Saba Khan
- 1Department of Biochemistry, Integral Institute of Medical Science and Research, Integral University, Lucknow, India
| |
Collapse
|
28
|
Khamoshina MB, Artemenko YS, Bayramova AA, Ryabova VA, Orazov MR. Polycystic ovary syndrome and obesity: a modern paradigm. RUDN JOURNAL OF MEDICINE 2022. [DOI: 10.22363/2313-0245-2022-26-4-382-395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Polycystic ovary syndrome is a heterogeneous endocrine disease that affects women of childbearing age. The pathogenesis of polycystic ovary syndrome has not been fully studied to date, its paradigm considers the genetic determinism of the manifestation of hormonal and metabolic disorders, which are considered to be criteria for the verification of the disease (hyperandrogenism, oligo/anovulation and/or polycystic ovarian transformation during ultrasound examination (ultrasound). This review discusses the main ways of interaction between hyperandrogenism, insulin resistance and obesity and their role in the pathogenesis of polycystic ovary syndrome, as well as possible methods of treatment for this category of patients. The review analyzes the role of hyperandrogenism and insulin resistance in the implementation of the genetic scenario of polycystic ovary syndrome and finds out the reasons why women with polycystic ovary syndrome often demonstrate the presence of a «metabolic trio» - hyperinsulinemia, insulin resistance and type 2 diabetes mellitus. It is noted that obesity is not included in the criteria for the diagnosis of polycystic ovary syndrome, but epidemiological data confirm the existence of a relationship between these diseases. Obesity, especially visceral, which is often found in women with polycystic ovary syndrome, enhances and worsens metabolic and reproductive outcomes with polycystic ovary syndrome, as well as increases insulin resistance and compensatory hyperinsulinemia, which, in turn, stimulates adipogenesis and suppresses lipolysis. Obesity increases the sensitivity of tech cells to luteinizing hormone stimulation and enhances functional hyperandrogenism of the ovaries, increasing the production of androgens by the ovaries. Excess body weight is associated with a large number of inflammatory adipokines, which, in turn, contribute to the growth of insulin resistance and adipogenesis. Obesity and insulin resistance exacerbate the symptoms of hyperandrogenism, forming a vicious circle that contributes to the development of polycystic ovary syndrome. These data allow us to conclude that bariatric surgery can become an alternative to drugs (metformin, thiazolidinedione analogs of glucagon-like peptide-1), which has shown positive results in the treatment of patients with polycystic ovary syndrome and obesity.
Collapse
|
29
|
Lempesis IG, Hoebers N, Essers Y, Jocken JWE, Rouschop KMA, Blaak EE, Manolopoulos KN, Goossens GH. Physiological Oxygen Levels Differentially Regulate Adipokine Production in Abdominal and Femoral Adipocytes from Individuals with Obesity Versus Normal Weight. Cells 2022; 11:cells11223532. [PMID: 36428961 PMCID: PMC9688196 DOI: 10.3390/cells11223532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022] Open
Abstract
Adipose tissue (AT) inflammation may increase obesity-related cardiometabolic complications. Altered AT oxygen partial pressure (pO2) may impact the adipocyte inflammatory phenotype. Here, we investigated the effects of physiological pO2 levels on the inflammatory phenotype of abdominal (ABD) and femoral (FEM) adipocytes derived from postmenopausal women with normal weight (NW) or obesity (OB). Biopsies were collected from ABD and FEM subcutaneous AT in eighteen postmenopausal women (aged 50-65 years) with NW (BMI 18-25 kg/m2, n = 9) or OB (BMI 30-40 kg/m2, n = 9). We compared the effects of prolonged exposure to different physiological pO2 levels on adipokine expression and secretion in differentiated human multipotent adipose-derived stem cells. Low physiological pO2 (5% O2) significantly increased leptin gene expression/secretion in ABD and FEM adipocytes derived from individuals with NW and OB compared with high physiological pO2 (10% O2) and standard laboratory conditions (21% O2). Gene expression/secretion of IL-6, DPP-4, and MCP-1 was reduced in differentiated ABD and FEM adipocytes from individuals with OB but not NW following exposure to low compared with high physiological pO2 levels. Low physiological pO2 decreases gene expression and secretion of several proinflammatory factors in ABD and FEM adipocytes derived from individuals with OB but not NW.
Collapse
Affiliation(s)
- Ioannis G. Lempesis
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
- Correspondence: (I.G.L.); (G.H.G.)
| | - Nicole Hoebers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Yvonne Essers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Johan W. E. Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Kasper M. A. Rouschop
- Radiotherapy, GROW School for Oncology & Reproduction, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Ellen E. Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Konstantinos N. Manolopoulos
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
- Correspondence: (I.G.L.); (G.H.G.)
| |
Collapse
|
30
|
Docosahexaenoic Acid Counteracts the Hypoxic-Induced Inflammatory and Metabolic Alterations in 3T3-L1 Adipocytes. Nutrients 2022; 14:nu14214600. [PMID: 36364860 PMCID: PMC9659308 DOI: 10.3390/nu14214600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/15/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
Background: Hypoxia is caused by the excessive expansion of the white adipose tissue (AT) and is associated with obesity-related conditions such as insulin resistance, inflammation, and oxidative stress. Docosahexaenoic acid (DHA) is an omega-3 fatty acid reported to have beneficial health effects. However, the effects of DHA in AT against hypoxia-induced immune-metabolic perturbations in adipocytes exposed to low O2 tension are not well known. Consequently, this study aimed to evaluate the impact of DHA on markers of inflammation, metabolism, apoptosis, and oxidative stress in 3T3-L1 cell adipocytes exposed to low O2 tension (1% O2) induced hypoxia. Methods: The apoptosis and reactive oxygen species (ROS) rates were evaluated. Metabolic parameters such as lactate, FFA, glycerol release, glucose uptake, and ATP content were assessed by a fluorometer. The expression of HIF-1, GLUT1 and the secretion of adipocytokines such as leptin, adiponectin, and pro-inflammatory markers was evaluated. Results: DHA-treated hypoxic cells showed significantly decreased basal free fatty acid release, lactate production, and enhanced glucose consumption. In addition, DHA-treatment of hypoxic cells caused a significant reduction in the apoptosis rate and ROS production with decreased lipid peroxidation. Moreover, DHA-treatment of hypoxic cells caused a decreased secretion of pro-inflammatory markers (IL-6, MCP-1) and leptin and increased adiponectin secretion compared with hypoxic cells. Furthermore, DHA-treatment of hypoxic cells caused significant reductions in the expression of genes related to hypoxia (HIF-1, HIF-2), anaerobic metabolism (GLUT1 and Ldha), ATP production (ANT2), and fat metabolism (FASN and PPARY). Conclusion: This study suggests that DHA can exert potential anti-obesity effects by reducing the secretion of inflammatory adipokines, oxidative stress, lipolysis, and apoptosis.
Collapse
|
31
|
Mika K, Szafarz M, Zadrożna M, Nowak B, Bednarski M, Szczepańska K, Pociecha K, Kubacka M, Nicosia N, Juda I, Kieć-Kononowicz K, Kotańska M. KSK-74: Dual Histamine H3 and Sigma-2 Receptor Ligand with Anti-Obesity Potential. Int J Mol Sci 2022; 23:ijms23137011. [PMID: 35806019 PMCID: PMC9266460 DOI: 10.3390/ijms23137011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 12/10/2022] Open
Abstract
Many studies involving compounds that enhance histamine release, such as histamine H3 receptor (H3R) antagonists, have shown efficacy in inhibiting weight gain, but none have passed clinical trials. As part of the search for H3 receptor ligands that have additional properties, the aim of this study is to evaluate the activity in the reduction in weight gain in a rat model of excessive eating, as well as the impact on selected metabolic parameters, and the number and size of adipocytes of two new H3R antagonists, KSK-60 and KSK-74, which also exert a significant affinity at the sigma-2 receptor. Compounds KSK-60 and KSK-74 are homologues and the elongation of the distal part of the molecule resulted in an approximate two-fold reduction in affinity at H3R, but simultaneously an almost two-fold increase in affinity at the sigma-2 receptor. Animals fed palatable feed and receiving KSK-60 or KSK-74 both at 10 mg/kg b.w. gained significantly less weight than animals in the control obese group. Moreover, KSK-74 significantly compensated for metabolic disturbances that accompany obesity, such as an increase in plasma triglyceride, resistin, and leptin levels; improved glucose tolerance; and protected experimental animals against adipocyte hypertrophy. Furthermore, KSK-74 inhibited the development of inflammation in obesity-exposed adipose tissue. The in vivo pharmacological activity of the tested ligands appears to correlate with the affinity at the sigma-2 receptors; however, the explanation of this phenomenon requires further and extended research.
Collapse
Affiliation(s)
- Kamil Mika
- Department of Pharmacological Screening, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (K.M.); (M.B.); (M.K.); (N.N.)
| | - Małgorzata Szafarz
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (M.S.); (K.P.)
| | - Monika Zadrożna
- Department of Cytobiology, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (M.Z.); (B.N.); (I.J.)
| | - Barbara Nowak
- Department of Cytobiology, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (M.Z.); (B.N.); (I.J.)
| | - Marek Bednarski
- Department of Pharmacological Screening, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (K.M.); (M.B.); (M.K.); (N.N.)
| | - Katarzyna Szczepańska
- Technology and Biotechnology of Medical Remedies, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (K.S.); (K.K.-K.)
- Department of Medicinal Chemistry, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna 12, 31-343 Cracow, Poland
| | - Krzysztof Pociecha
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (M.S.); (K.P.)
| | - Monika Kubacka
- Department of Pharmacological Screening, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (K.M.); (M.B.); (M.K.); (N.N.)
| | - Noemi Nicosia
- Department of Pharmacological Screening, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (K.M.); (M.B.); (M.K.); (N.N.)
- Foundation “Prof. Antonio Imbesi”, University of Messina, Piazza Pugliatti 1, 98122 Messina, Italy
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Palatucci, 98168 Messina, Italy
| | - Izabela Juda
- Department of Cytobiology, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (M.Z.); (B.N.); (I.J.)
| | - Katarzyna Kieć-Kononowicz
- Technology and Biotechnology of Medical Remedies, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (K.S.); (K.K.-K.)
| | - Magdalena Kotańska
- Department of Pharmacological Screening, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland; (K.M.); (M.B.); (M.K.); (N.N.)
- Correspondence: ; Tel./Fax: +48-12-6205530
| |
Collapse
|
32
|
Salman AA, Salman MA, Said M, El Sherbiny M, Elkassar H, Hassan MB, Marwan A, Morad MA, Ashoush O, Labib S, Aon MH, Awad A, Sayed M, Taha AE, Moustafa A, Shaaban HED, Khater A, Elewa A, Khalaf AM, Mostafa AA, Matter M, Youssef A. Improvement of Pancreatic Steatosis and Indices of Insulin Resistance After Metabolic Surgery. Front Med (Lausanne) 2022; 9:894465. [PMID: 35733870 PMCID: PMC9207952 DOI: 10.3389/fmed.2022.894465] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose Obesity is associated with fat accumulation in ectopic sites such as the pancreas, the so-called pancreatic steatosis (PS). Bariatric surgery has been shown to be associated with reducing pancreatic fat. This study investigated the effect of laparoscopic sleeve gastrectomy (LSG) on pancreatic volume and its fat content and glucose homeostasis. Methods The study enrolled 54 patients subjected to LSG. Metabolic variables and pancreatic exocrine function were assessed immediately before surgery and 12 months after. MRI of the abdomen was performed to measure pancreatic fat content and its total volume and visceral adipose tissue (VAT). Results Surgery resulted in a significant reduction in body weight and BMI. HbA1c, fasting insulin, C-peptide levels, HOMA-IR, and Hs-CRP levels decreased significantly. Surgery resulted in significant improvement in lipid profile except for HDL-cholesterol and liver function tests. Total VAT volume decreased significantly. Total pancreas volume decreased by a mean of 9.0 cm3 (95% CI: 6.6–11.3). The median change of pancreatic fat was −26.1% (range: −55.6 to 58.3%). Pancreatic lipase decreased significantly (P < 0.001). There was a positive correlation between the percentage of total weight loss and decrease in pancreatic fat volume (r = 0.295, P = 0.030). Conclusion Weight loss after LSG is associated with a reduction of total VAT volume, total pancreatic volume, and pancreatic fat content. These changes are associated with improved glucose homeostasis, reduced systemic inflammation, and decreased pancreatic lipase secretion.
Collapse
Affiliation(s)
- Ahmed Abdallah Salman
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
- *Correspondence: Ahmed Abdallah Salman
| | | | - Mostafa Said
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohammad El Sherbiny
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hesham Elkassar
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Badr Hassan
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ahmed Marwan
- Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | - Omar Ashoush
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Safa Labib
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed H. Aon
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Abeer Awad
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Sayed
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ahmed E. Taha
- Department of Endemic Medicine and Hepatology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ahmed Moustafa
- Department of Endemic Medicine and Hepatology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hossam El-Din Shaaban
- Tropical and Gastroenterology Department, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Amir Khater
- Tropical and Gastroenterology Department, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Ahmed Elewa
- General Surgery Department, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Adel M. Khalaf
- Department of General Surgery, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Ahmed A. Mostafa
- Department of General Surgery, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Mohamed Matter
- Radiodiagnosis Department, Faculty of Medicine, Alazhar University, Cairo, Egypt
| | - Ahmed Youssef
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
33
|
Taghizadeh S, Gholizadeh M, Rahimi-Mianji G, Moradi MH, Costilla R, Moore S, Di Gerlando R. Genome-wide identification of copy number variation and association with fat deposition in thin and fat-tailed sheep breeds. Sci Rep 2022; 12:8834. [PMID: 35614300 PMCID: PMC9132911 DOI: 10.1038/s41598-022-12778-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 05/10/2022] [Indexed: 12/12/2022] Open
Abstract
Copy number variants (CNVs) are a type of genetic polymorphism which contribute to phenotypic variation in several species, including livestock. In this study, we used genomic data of 192 animals from 3 Iranian sheep breeds including 96 Baluchi sheep and 47 Lori-Bakhtiari sheep as fat-tailed breeds and 47 Zel sheep as thin-tailed sheep breed genotyped with Illumina OvineSNP50K Beadchip arrays. Also, for association test, 70 samples of Valle del Belice sheep were added to the association test as thin-tailed sheep breed. PennCNV and CNVRuler software were, respectively, used to study the copy number variation and genomic association analyses. We detected 573 and 242 CNVs in the fat and thin tailed breeds, respectively. In terms of CNV regions (CNVRs), these represented 328 and 187 CNVRs that were within or overlapping with 790 known Ovine genes. The CNVRs covered approximately 73.85 Mb of the sheep genome with average length 146.88 kb, and corresponded to 2.6% of the autosomal genome sequence. Five CNVRs were randomly chosen for validation, of which 4 were experimentally confirmed using Real time qPCR. Functional enrichment analysis showed that genes harbouring CNVs in thin-tailed sheep were involved in the adaptive immune response, regulation of reactive oxygen species biosynthetic process and response to starvation. In fat-tailed breeds these genes were involved in cellular protein modification process, regulation of heart rate, intestinal absorption, olfactory receptor activity and ATP binding. Association test identified one copy gained CNVR on chromosomes 6 harbouring two protein-coding genes HGFAC and LRPAP1. Our findings provide information about genomic structural changes and their association to the interested traits including fat deposition and environmental compatibility in sheep.
Collapse
Affiliation(s)
- Shadan Taghizadeh
- Department of Animal Science, Faculty of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University, P.O. Box - 578, Sari, Iran
| | - Mohsen Gholizadeh
- Department of Animal Science, Faculty of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University, P.O. Box - 578, Sari, Iran.
| | - Ghodrat Rahimi-Mianji
- Department of Animal Science, Faculty of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University, P.O. Box - 578, Sari, Iran
| | - Mohammad Hossein Moradi
- Department of Animal Science, Faculty of Agriculture and Natural Resources, Arak University, Arak, Iran
| | - Roy Costilla
- Ruakura Research Centre, AgResearch, Hamilton, New Zealand
| | - Stephen Moore
- Queensland Alliance for Agriculture and Food Innovation, University of Queensland, Brisbane, Australia
| | - Rosalia Di Gerlando
- Dipartimento Di Scienze Agrarie, Alimentari E Forestali, Università Degli Studi Di Palermo, Palermo, Italy
| |
Collapse
|
34
|
Yin R, Xu Y, Wang X, Yang L, Zhao D. Role of Dipeptidyl Peptidase 4 Inhibitors in Antidiabetic Treatment. Molecules 2022; 27:3055. [PMID: 35630534 PMCID: PMC9147686 DOI: 10.3390/molecules27103055] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/02/2022] [Accepted: 05/07/2022] [Indexed: 02/07/2023] Open
Abstract
In recent years, important changes have occurred in the field of diabetes treatment. The focus of the treatment of diabetic patients has shifted from the control of blood glucose itself to the overall management of risk factors, while adjusting blood glucose goals according to individualization. In addition, regulators need to approve new antidiabetic drugs which have been tested for cardiovascular safety. Thus, the newest class of drugs has been shown to reduce major adverse cardiovascular events, including sodium-glucose transporter 2 (SGLT2) and some glucagon like peptide 1 receptor (GLP1) analog. As such, they have a prominent place in the hyperglycemia treatment algorithms. In recent years, the role of DPP4 inhibitors (DPP4i) has been modified. DPP4i have a favorable safety profile and anti-inflammatory profile, do not cause hypoglycemia or weight gain, and do not require dose escalation. In addition, it can also be applied to some types of chronic kidney disease patients and elderly patients with diabetes. Overall, DPP4i, as a class of safe oral hypoglycemic agents, have a role in the management of diabetic patients, and there is extensive experience in their use.
Collapse
Affiliation(s)
| | | | | | | | - Dong Zhao
- Beijing Key Laboratory of Diabetes Prevention and Research, Center for Endocrine Metabolic and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China; (R.Y.); (Y.X.); (X.W.); (L.Y.)
| |
Collapse
|
35
|
Moreira VC, Silva CMS, Welker AF, da Silva ICR. Visceral Adipose Tissue Influence on Health Problem Development and Its Relationship with Serum Biochemical Parameters in Middle-Aged and Older Adults: A Literature Review. J Aging Res 2022; 2022:8350527. [PMID: 35492380 PMCID: PMC9042620 DOI: 10.1155/2022/8350527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/28/2021] [Accepted: 03/29/2022] [Indexed: 11/24/2022] Open
Abstract
Background The amount of visceral adipose tissue (VAT) tends to increase with age and is associated with several health problems, such as cardiometabolic diseases, increased infections, and overall mortality. Objectives This review provides a general assessment of how visceral adiposity correlates with the development of health problems and changes in serum biochemical parameters in middle-aged and older adults. Methods We searched specific terms in the Virtual Health Library (VHL) databases for VAT articles published in the English language between 2009 and 2019 related to older adults. Results The search found twenty-three publications in this period, of which nine were excluded. The publications had a population aged between 42 and 83 years and correlated the VAT area ratio with several comorbidities (such as pancreatitis, depression, cancer, and coronary heart disease) and serum biochemical parameters. Conclusion Further research on the association between visceral obesity and the emergence of health problems and the relationship between VAT and changes in serum biochemical parameters in older individuals should deepen the understanding of this connection and develop preventive actions.
Collapse
Affiliation(s)
- Vanessa C. Moreira
- Health Sciences and Technologies, University of Brasilia, Zip-Code: 72220-275, Brasilia, Brazil
| | - Calliandra M. S. Silva
- Health Sciences and Technologies, University of Brasilia, Zip-Code: 72220-275, Brasilia, Brazil
| | - Alexis F. Welker
- Health Sciences and Technologies, University of Brasilia, Zip-Code: 72220-275, Brasilia, Brazil
| | - Izabel C. R. da Silva
- Health Sciences and Technologies, University of Brasilia, Zip-Code: 72220-275, Brasilia, Brazil
| |
Collapse
|
36
|
Pitere RR, van Heerden MB, Pepper MS, Ambele MA. Slc7a8 Deletion Is Protective against Diet-Induced Obesity and Attenuates Lipid Accumulation in Multiple Organs. BIOLOGY 2022; 11:311. [PMID: 35205177 PMCID: PMC8869389 DOI: 10.3390/biology11020311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 11/22/2022]
Abstract
Adipogenesis, through adipocyte hyperplasia and/or hypertrophy, leads to increased adiposity, giving rise to obesity. A genome-wide transcriptome analysis of in vitro adipogenesis in human adipose-derived stromal/stem cells identified SLC7A8 (Solute Carrier Family 7 Member 8) as a potential novel mediator. The current study has investigated the role of SLC7A8 in adipose tissue biology using a mouse model of diet-induced obesity. slc7a8 knockout (KO) and wildtype (WT) C57BL/6J mice were fed either a control diet (CD) or a high-fat diet (HFD) for 14 weeks. On the HFD, both WT and KO mice (WTHFD and KOHFD) gained significantly more weight than their CD counterparts. However, KOHFD gained significantly less weight than WTHFD. KOHFD had significantly reduced levels of glucose intolerance compared with those observed in WTHFD. KOHFD also had significantly reduced adipocyte mass and hypertrophy in inguinal, mesenteric, perigonadal, and brown adipose depots, with a corresponding decrease in macrophage infiltration. Additionally, KOHFD had decreased lipid accumulation in the liver, heart, gastrocnemius muscle, lung, and kidney. This study demonstrates that targeting slc7a8 protects against diet-induced obesity by reducing lipid accumulation in multiple organs and suggests that if targeted, has the potential to mitigate the development of obesity-associated comorbidities.
Collapse
Affiliation(s)
- Reabetswe R. Pitere
- Institute for Cellular and Molecular Medicine, Department of Immunology and SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (R.R.P.); (M.S.P.)
| | - Marlene B. van Heerden
- Department of Oral Pathology and Oral Biology, School of Dentistry, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology and SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (R.R.P.); (M.S.P.)
| | - Melvin A. Ambele
- Institute for Cellular and Molecular Medicine, Department of Immunology and SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (R.R.P.); (M.S.P.)
- Department of Oral Pathology and Oral Biology, School of Dentistry, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| |
Collapse
|
37
|
Wang M, Ma W, Wang Q, Yang Q, Yan X, Tang H, Li Z, Li Y, Feng S, Wang Z. Flavonoid-enriched extract from Millettia speciosa Champ prevents obesity by regulating thermogenesis and lipid metabolism in high-fat diet-induced obese C57BL/6 mice. Food Sci Nutr 2022; 10:445-459. [PMID: 35154681 PMCID: PMC8825741 DOI: 10.1002/fsn3.2664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/13/2021] [Accepted: 10/27/2021] [Indexed: 01/01/2023] Open
Abstract
Millettia speciosa (M. speciosa) Champ is a medicinal and edible plant. The roots are rich in flavonoids, which possess multiple biological activities, including lipid-lowering effects. This study aimed to explore the effect of flavonoid-enriched extract from M. speciosa (FMS) on obesity. The UPLC-Q-TOF-MS analysis and chromatographic analysis were adopted to identify flavonoid compounds in FMS. Male C57BL/6J mice were fed with a high-fat diet for 3 months and were then treated with FMS (50 or 100 mg/kg/d) or Orlistat (10 mg kg-1 d-1) for another 8 weeks. A total of 35 flavonoids were identified in the extract of M. speciosa root. FMS reduced body weight gain, liver weight gain, white adipose tissue, lipid accumulation, and blood glucose. The levels of TG, ALT, AST, and inflammatory-related adipokines (TNF-α and IL-6) in serum were also reduced by FMS. In addition, FMS promoted thermogenesis in brown adipose tissue and induced the activation of lipolysis, fatty acid oxidation, and oxidative phosphorylation in white adipose tissues. In summary, long-term administration of FMS could ameliorate high-fat diet-induced obesity by stimulating adipose thermogenesis and lipid metabolism.
Collapse
Affiliation(s)
- Mao‐Yuan Wang
- Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern ChinaTropical Crops Genetic Resources InstituteMinistry of AgricultureHaikouChina
- Tropical Wild Plant Gene ResourceMinistry of Agriculture/National Genebank of Tropical CropsDanzhouChina
| | - Wen‐Yu Ma
- Key Laboratory of South Subtropical Plant DiversityFairy Lake Botanical GardenShenzhen & Chinese Academy of SciencesShenzhenChina
| | - Qing‐Long Wang
- Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern ChinaTropical Crops Genetic Resources InstituteMinistry of AgricultureHaikouChina
- Tropical Wild Plant Gene ResourceMinistry of Agriculture/National Genebank of Tropical CropsDanzhouChina
| | - Qing Yang
- Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern ChinaTropical Crops Genetic Resources InstituteMinistry of AgricultureHaikouChina
- Tropical Wild Plant Gene ResourceMinistry of Agriculture/National Genebank of Tropical CropsDanzhouChina
| | - Xiao‐Xia Yan
- Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern ChinaTropical Crops Genetic Resources InstituteMinistry of AgricultureHaikouChina
- Tropical Wild Plant Gene ResourceMinistry of Agriculture/National Genebank of Tropical CropsDanzhouChina
| | - Huan Tang
- Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern ChinaTropical Crops Genetic Resources InstituteMinistry of AgricultureHaikouChina
- Tropical Wild Plant Gene ResourceMinistry of Agriculture/National Genebank of Tropical CropsDanzhouChina
| | - Zhi‐Ying Li
- Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern ChinaTropical Crops Genetic Resources InstituteMinistry of AgricultureHaikouChina
- Tropical Wild Plant Gene ResourceMinistry of Agriculture/National Genebank of Tropical CropsDanzhouChina
| | - Ying‐Ying Li
- Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern ChinaTropical Crops Genetic Resources InstituteMinistry of AgricultureHaikouChina
- Tropical Wild Plant Gene ResourceMinistry of Agriculture/National Genebank of Tropical CropsDanzhouChina
| | - Shi‐Xiu Feng
- Key Laboratory of South Subtropical Plant DiversityFairy Lake Botanical GardenShenzhen & Chinese Academy of SciencesShenzhenChina
| | - Zhu‐Nian Wang
- Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern ChinaTropical Crops Genetic Resources InstituteMinistry of AgricultureHaikouChina
- Tropical Wild Plant Gene ResourceMinistry of Agriculture/National Genebank of Tropical CropsDanzhouChina
| |
Collapse
|
38
|
Clément AA, Lacaille M, Lounis MA, Biertho LD, Richard D, Lemieux I, Bergeron J, Mounier C, Joanisse DR, Mauriège P. Intra-abdominal adipose depot variation in adipogenesis, lipogenesis, angiogenesis, and fibrosis gene expression and relationships with insulin resistance and inflammation in premenopausal women with severe obesity. J Physiol Biochem 2022; 78:527-542. [PMID: 35000091 DOI: 10.1007/s13105-021-00855-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/21/2021] [Indexed: 10/19/2022]
Abstract
Although severe obesity is associated with insulin resistance (IR) and inflammation, secretory function of intra-abdominal adipose tissues and their relationships with IR and inflammation markers remain poorly understood. Aims were to measure gene expression of adipogenic (C/EBPα/β, PPARγ-1/2, SREBP-1c, LXRα), lipogenic (SCD1, DGAT-1/2), angiogenic (VEGFα, leptin), and fibrotic (LOX, COL6A3) factors in the round ligament (RL), omental (OM), and mesenteric (ME) fat depots and to evaluate their relationships with IR and inflammation markers in 48 women with severe obesity undergoing bariatric surgery. Gene expression was assessed by RT-qPCR, and plasma glucose and insulin (HOMA-IR calculated), PAI-1, IL-6, TNFα, adiponectin, and leptin levels were determined. C/EBPβ and PPARγ-1/2 mRNA levels were more expressed in the OM (0.001<p<0.05). ME showed the highest expression of C/EBPα, SREBP-1c, DGAT-2, and leptin and the lowest of SCD1, LXRα, VEGFα, and LOX (0.001<p<0.05). COL6A3 expression was higher in the ME and RL (p<0.001). COL6A3 expression was negatively associated with IR indices in the RL (0.01<p<0.05) and with fasting glycemia and HOMA-IR in the OM (0.001<p<0.05). VEGFα expression was positively related to TNFα and PAI-1 in the RL (0.001<p<0.05) and to PAI-1 in the OM (p<0.05) and negatively to PAI-1 in the ME (p<0.001). Fibrosis gene expression correlated negatively with inflammation in RL and OM (0.001<p<0.05) and positively with PAI-1 in the ME (0.001<p<0.05). The varying relationships of gene expression profiles with selected IR indices and inflammation biomarkers further suggest these fat depots have distinct contributions to overall health in premenopausal women with severe obesity.
Collapse
Affiliation(s)
- Andrée-Anne Clément
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec (CRIUCPQ), Quebec, Canada.,Département de Biochimie et Génomique Fonctionnelle, Faculté de médecine et sciences de la santé, Université de Sherbrooke, Sherbrooke, Canada
| | - Michel Lacaille
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec (CRIUCPQ), Quebec, Canada
| | - Mohamed Amine Lounis
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Institut du Cancer de Montréal (ICM), Montreal, Quebec, Canada
| | - Laurent D Biertho
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec (CRIUCPQ), Quebec, Canada.,Département de Chirurgie, Faculté de Médecine, Université Laval, Quebec, Canada
| | - Denis Richard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec (CRIUCPQ), Quebec, Canada.,Département de Physiologie, Faculté de Médecine, Université Laval, Quebec, Canada
| | - Isabelle Lemieux
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec (CRIUCPQ), Quebec, Canada
| | - Jean Bergeron
- Axe Endocrinologie et Néphrologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec (CHUL), Québec, Canada
| | - Catherine Mounier
- Département des Sciences Biologiques et Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec À Montréal, Montreal, Canada
| | - Denis R Joanisse
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec (CRIUCPQ), Quebec, Canada.,Département de Kinésiologie, Faculté de médecine, Université Laval, Quebec, Canada
| | - Pascale Mauriège
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec (CRIUCPQ), Quebec, Canada. .,Département de Kinésiologie, Faculté de médecine, Université Laval, Quebec, Canada.
| |
Collapse
|
39
|
Tan Q, Akindehin SE, Orsso CE, Waldner RC, DiMarchi RD, Müller TD, Haqq AM. Recent Advances in Incretin-Based Pharmacotherapies for the Treatment of Obesity and Diabetes. Front Endocrinol (Lausanne) 2022; 13:838410. [PMID: 35299971 PMCID: PMC8921987 DOI: 10.3389/fendo.2022.838410] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/07/2022] [Indexed: 01/01/2023] Open
Abstract
The incretin hormone glucagon-like peptide-1 (GLP-1) has received enormous attention during the past three decades as a therapeutic target for the treatment of obesity and type 2 diabetes. Continuous improvement of the pharmacokinetic profile of GLP-1R agonists, starting from native hormone with a half-life of ~2-3 min to the development of twice daily, daily and even once-weekly drugs highlight the pharmaceutical evolution of GLP-1-based medicines. In contrast to GLP-1, the incretin hormone glucose-dependent insulinotropic polypeptide (GIP) received little attention as a pharmacological target, because of conflicting observations that argue activation or inhibition of the GIP receptor (GIPR) provides beneficial effects on systemic metabolism. Interest in GIPR agonism for the treatment of obesity and diabetes was recently propelled by the clinical success of unimolecular dual-agonists targeting the receptors for GIP and GLP-1, with reported significantly improved body weight and glucose control in patients with obesity and type II diabetes. Here we review the biology and pharmacology of GLP-1 and GIP and discuss recent advances in incretin-based pharmacotherapies.
Collapse
Affiliation(s)
- Qiming Tan
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Seun E. Akindehin
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Germany and German Center for Diabetes Research (DZD), Munich, Germany
| | - Camila E. Orsso
- Department of Agricultural Food & Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | | | | | - Timo D. Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Germany and German Center for Diabetes Research (DZD), Munich, Germany
- *Correspondence: Timo D. Müller, ; Andrea M. Haqq,
| | - Andrea M. Haqq
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
- Department of Agricultural Food & Nutritional Science, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Timo D. Müller, ; Andrea M. Haqq,
| |
Collapse
|
40
|
Sagris M, Theofilis P, Antonopoulos AS, Oikonomou E, Paschaliori C, Galiatsatos N, Tsioufis K, Tousoulis D. Inflammation in Coronary Microvascular Dysfunction. Int J Mol Sci 2021; 22:ijms222413471. [PMID: 34948272 PMCID: PMC8703507 DOI: 10.3390/ijms222413471] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/05/2021] [Accepted: 12/07/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic low-grade inflammation is involved in coronary atherosclerosis, presenting multiple clinical manifestations ranging from asymptomatic to stable angina, acute coronary syndrome, heart failure and sudden cardiac death. Coronary microvasculature consists of vessels with a diameter less than 500 μm, whose potential structural and functional abnormalities can lead to inappropriate dilatation and an inability to meet the required myocardium oxygen demands. This review focuses on the pathogenesis of coronary microvascular dysfunction and the capability of non-invasive screening methods to detect the phenomenon. Anti-inflammatory agents, such as statins and immunomodulators, including anakinra, tocilizumab, and tumor necrosis factor-alpha inhibitors, have been assessed recently and may constitute additional or alternative treatment approaches to reduce cardiovascular events in atherosclerotic heart disease characterized by coronary microvascular dysfunction.
Collapse
Affiliation(s)
- Marios Sagris
- Cardiology Clinic, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (C.P.); (N.G.); (K.T.); (D.T.)
- Correspondence: ; Tel.:+30-213-2088099; Fax: +30-213-2088676
| | - Panagiotis Theofilis
- Cardiology Clinic, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (C.P.); (N.G.); (K.T.); (D.T.)
| | - Alexios S. Antonopoulos
- Cardiology Clinic, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (C.P.); (N.G.); (K.T.); (D.T.)
| | - Evangelos Oikonomou
- Cardiology Clinic, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (C.P.); (N.G.); (K.T.); (D.T.)
- Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, University of Athens Medical School, 11527 Athens, Greece
| | - Christina Paschaliori
- Cardiology Clinic, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (C.P.); (N.G.); (K.T.); (D.T.)
| | - Nikolaos Galiatsatos
- Cardiology Clinic, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (C.P.); (N.G.); (K.T.); (D.T.)
| | - Kostas Tsioufis
- Cardiology Clinic, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (C.P.); (N.G.); (K.T.); (D.T.)
| | - Dimitris Tousoulis
- Cardiology Clinic, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (C.P.); (N.G.); (K.T.); (D.T.)
| |
Collapse
|
41
|
McColloch A, Liu H, Cho M. Reversal of stem cell‐derived hypertrophic adipocytes mediated by photobiomodulation (1064 nm). TRANSLATIONAL BIOPHOTONICS 2021. [DOI: 10.1002/tbio.202100006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Andrew McColloch
- Department of Bioengineering The University of Texas at Arlington Arlington Texas USA
| | - Hanli Liu
- Department of Bioengineering The University of Texas at Arlington Arlington Texas USA
| | - Michael Cho
- Department of Bioengineering The University of Texas at Arlington Arlington Texas USA
| |
Collapse
|
42
|
Corrales P, Vidal-Puig A, Medina-Gómez G. Obesity and pregnancy, the perfect metabolic storm. Eur J Clin Nutr 2021; 75:1723-1734. [PMID: 33911209 DOI: 10.1038/s41430-021-00914-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/15/2021] [Accepted: 03/29/2021] [Indexed: 02/02/2023]
Abstract
Pregnancy is a physiological stress that requires dynamic, regulated changes affecting maternal and fetal adiposity. Excessive accumulation of dysfunctional adipose tissue defined by metabolic and molecular alterations cause severe health consequences for mother and fetus. When subjected to sustained overnutrition, the cellular and lipid composition of the adipose tissue changes predisposing to insulin resistance, diabetes, and other metabolic disorders compromising the outcome of the pregnancy. Moreover, excessive maternal weight gain, usually in the context of obesity, predisposes to an increased flux of nutrients from mother to fetus throughout the placenta. The fetus of an obese mother will accumulate more adiposity and may increase the risk of future metabolic disorder later in life. Thus, further understanding of the interaction between maternal metabolism, epigenetic regulation of the adipose tissue, and their transgenerational transfer are required to mitigate the adverse health outcomes for the mother and the fetus associated with maternal obesity.
Collapse
Affiliation(s)
- Patricia Corrales
- Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Spain.
| | - Antonio Vidal-Puig
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, UK
- Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, PR China
| | - Gema Medina-Gómez
- Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Spain.
| |
Collapse
|
43
|
Mattioli AV, Coppi F, Manenti A, Farinetti A. Subclinical Vascular Damage: Current Insights and Future Potential. Vasc Health Risk Manag 2021; 17:729-738. [PMID: 34866906 PMCID: PMC8633547 DOI: 10.2147/vhrm.s242636] [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: 09/03/2021] [Accepted: 11/13/2021] [Indexed: 12/24/2022] Open
Abstract
The cardiovascular risk assessment must be carried out during all the different phases of life because the cardiovascular risk and the related prevention actions are dynamic and constantly evolving. As patients age, they change their exposure to various risk factors and accumulate comorbidities by changing their subjective cardiovascular risk, so it is necessary to undertake personalized early and preventive diagnostic actions. The main approach to asymptomatic vascular disease is based on primary prevention with the adoption of a healthy lifestyle. Indeed, lifestyle influences most of the traditional risk factors. In recent years, important differences between the sexes regarding cardiovascular risk factors have emerged and in particular, risk factors specific for female sex have been identified. Women are more likely to be categorized into lower risk categories for cardiovascular disease and, as a result, receive less lifestyle counseling than men, as well as less intensive prevention. This narrative review aims to analyze CVD risk prevention in asymptomatic atherosclerosis with a look at new emerging factors. In the end, we quickly analyzed the effects of the recent pandemic on lifestyle and cardiovascular risk and the potential negative effects in the long term.
Collapse
Affiliation(s)
- Anna Vittoria Mattioli
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Antonio Manenti
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Alberto Farinetti
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
44
|
Reavey JJ, Walker C, Murray AA, Brito-Mutunayagam S, Sweeney S, Nicol M, Cambursano A, Critchley HOD, Maybin JA. Obesity is associated with heavy menstruation that may be due to delayed endometrial repair. J Endocrinol 2021; 249:71-82. [PMID: 33836495 PMCID: PMC8052524 DOI: 10.1530/joe-20-0446] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/09/2021] [Indexed: 12/22/2022]
Abstract
Heavy menstrual bleeding is common and debilitating but the causes remain ill defined. Rates of obesity in women are increasing and its impact on menstrual blood loss (MBL) is unknown. Therefore, we quantified BMI and MBL in women not taking hormones and with regular menstrual cycles and revealed a positive correlation. In a mouse model of simulated menstruation, diet-induced obesity also resulted in delayed endometrial repair, a surrogate marker for MBL. BrdU staining of mouse uterine tissue revealed decreased proliferation during menstruation in the luminal epithelium of mice on a high-fat diet. Menstruation is known to initiate local endometrial inflammation and endometrial hypoxia; hence, the impact of body weight on these processes was investigated. A panel of hypoxia-regulated genes (VEGF, ADM, LDHA, SLC2A1) showed consistently higher mean values in the endometrium of women with obesity and in uteri of mice with increased weight vs normal controls, although statistical significance was not reached. The inflammatory mediators, Tnf and Il6 were significantly increased in the uterus of mice on a high-fat diet, consistent with a pro-inflammatory local endometrial environment in these mice. In conclusion, obesity was associated with increased MBL in women. Mice given a high-fat diet had delayed endometrial repair at menstruation and provided a model in which to study the influence of obesity on menstrual physiology. Our results indicate that obesity results in a more pro-inflammatory local endometrial environment at menstruation, which may delay endometrial repair and increase menstrual blood loss.
Collapse
Affiliation(s)
- Jane J Reavey
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Catherine Walker
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Alison A Murray
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | | | - Sheona Sweeney
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Moira Nicol
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Ana Cambursano
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | | | | |
Collapse
|
45
|
Atakan MM, Koşar ŞN, Güzel Y, Tin HT, Yan X. The Role of Exercise, Diet, and Cytokines in Preventing Obesity and Improving Adipose Tissue. Nutrients 2021; 13:nu13051459. [PMID: 33922998 PMCID: PMC8145589 DOI: 10.3390/nu13051459] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/17/2022] Open
Abstract
The prevalence of obesity continues to rise worldwide despite evidence-based public health recommendations. The promise to adopt a healthy lifestyle is increasingly important for tackling this global epidemic. Calorie restriction or regular exercise or a combination of the two is accepted as an effective strategy in preventing or treating obesity. Furthermore, the benefits conferred by regular exercise to overcome obesity are attributed not only to reduced adiposity or reduced levels of circulating lipids but also to the proteins, peptides, enzymes, and metabolites that are released from contracting skeletal muscle or other organs. The secretion of these molecules called cytokines in response to exercise induces browning of white adipose tissue by increasing the expression of brown adipocyte-specific genes within the white adipose tissue, suggesting that exercise-induced cytokines may play a significant role in preventing obesity. In this review, we present research-based evidence supporting the effects of exercise and various diet interventions on preventing obesity and adipose tissue health. We also discuss the interplay between adipose tissue and the cytokines secreted from skeletal muscle and other organs that are known to affect adipose tissue and metabolism.
Collapse
Affiliation(s)
- Muhammed Mustafa Atakan
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey; (M.M.A.); (Ş.N.K.); (Y.G.)
| | - Şükran Nazan Koşar
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey; (M.M.A.); (Ş.N.K.); (Y.G.)
| | - Yasemin Güzel
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey; (M.M.A.); (Ş.N.K.); (Y.G.)
| | - Hiu Tung Tin
- Institute for Health and Sport (iHeS), Victoria University, P.O. Box 14428, Melbourne 8001, Australia;
| | - Xu Yan
- Institute for Health and Sport (iHeS), Victoria University, P.O. Box 14428, Melbourne 8001, Australia;
- Sarcopenia Research Program, Australia Institute for Musculoskeletal Sciences (AIMSS), Melbourne 3021, Australia
- Correspondence: ; Tel.: +61-3-9919-4024; Fax: +61-3-9919-5615
| |
Collapse
|
46
|
Suvarnapathaki S, Nguyen MA, Goulopoulos AA, Lantigua D, Camci-Unal G. Engineering calcium peroxide based oxygen generating scaffolds for tissue survival. Biomater Sci 2021; 9:2519-2532. [PMID: 33565527 PMCID: PMC11442008 DOI: 10.1039/d0bm02048f] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Oxygen supply is essential for the long-term viability and function of tissue engineered constructs in vitro and in vivo. The integration with the host blood supply as the primary source of oxygen to cells requires 4 to 5 weeks in vivo and involves neovascularization stages to support the delivery of oxygenated blood to cells. Consequently, three-dimensional (3D) encapsulated cells during this process are prone to oxygen deprivation, cellular dysfunction, damage, and hypoxia-induced necrosis. Here we demonstrate the use of calcium peroxide (CaO2) and polycaprolactone (PCL), as part of an emerging paradigm of oxygen-generating scaffolds that substitute the host oxygen supply via hydrolytic degradation. The 35-day in vitro study showed predictable oxygen release kinetics that achieved 5% to 29% dissolved oxygen with increasing CaO2 loading. As a biomaterial, the iterations of 0 mg, 40 mg, and 60 mg of CaO2 loaded scaffolds yielded modular mechanical behaviors, ranging from 5-20 kPa in compressive strength. The other controlled physiochemical features included swelling capacities of 22-33% and enzymatic degradation rates of 0.8% to 60% remaining mass. The 3D-encapsulation experiments of NIH/3T3 fibroblasts, L6 rat myoblasts, and primary cardiac fibroblasts in these scaffolds showed enhanced cell survival, proliferation, and function under hypoxia. During continuous oxygen release, the scaffolds maintained a stable tissue culture system between pH 8 to 9. The broad basis of this work supports prospects in the expansion of robust and clinically translatable tissue constructs.
Collapse
Affiliation(s)
- Sanika Suvarnapathaki
- Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell, One University Avenue, Lowell, MA 01854, USA
| | | | | | | | | |
Collapse
|
47
|
Moruzzi M, Klöting N, Blüher M, Martinelli I, Tayebati SK, Gabrielli MG, Roy P, Micioni Di Bonaventura MV, Cifani C, Lupidi G, Amenta F, Tomassoni D. Tart Cherry Juice and Seeds Affect Pro-Inflammatory Markers in Visceral Adipose Tissue of High-Fat Diet Obese Rats. Molecules 2021; 26:1403. [PMID: 33807712 PMCID: PMC7961347 DOI: 10.3390/molecules26051403] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/03/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Tart cherries (Prunus cerasus L.) are a rich source of anthocyanins. They are phytochemical flavonoids found in red and blue fruits, and vegetables that can reduce hyperlipidemia. Visceral Adipose Tissue (VAT) has emerged as a major player in driving obesity-related inflammatory response. METHODS This study has investigated the potential positive effects of tart cherries on rats with Diet-Induced Obesity (DIO). In particular, the inflammatory status in retroperitoneal (RPW) and perigonadal (PGW) adipose tissue were studied. Rats were fed ad libitum for 17 weeks with a hypercaloric diet with the supplementation of tart cherries seeds powder (DS) and seeds powder plus tart cherries juice containing 1mg of anthocyanins (DJS). In RPW and PGW, expression of CRP, IL-1 β, TNF-α, CCL2 and CD36, were measured by qRT-PCR, Western blot and immunohistochemistry techniques. RESULTS No differences in the weight of RPW and PGW animals were found between DS and DJS groups compared to DIO rats. However, an increase of inflammatory markers was observed in DIO group in comparison with control lean rats. A modulation of these markers was evident upon tart cherry supplementation. CONCLUSION Study results suggest that tart cherry enriched-diet did not modify the accumulation of visceral fat, but it decreased inflammatory markers in both tissues. Therefore, this supplementation could be useful, in combination with healthy lifestyles, to modify adipose tissue cell metabolism limiting-obesity related organ damage.
Collapse
Affiliation(s)
- Michele Moruzzi
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.M.); (I.M.); (S.K.T.); (M.V.M.D.B.); (C.C.); (G.L.); (F.A.)
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany; (N.K.); (M.B.)
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany; (N.K.); (M.B.)
| | - Ilenia Martinelli
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.M.); (I.M.); (S.K.T.); (M.V.M.D.B.); (C.C.); (G.L.); (F.A.)
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, 31432 Toulouse, France
| | - Seyed Khosrow Tayebati
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.M.); (I.M.); (S.K.T.); (M.V.M.D.B.); (C.C.); (G.L.); (F.A.)
| | - Maria Gabriella Gabrielli
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (M.G.G.); (P.R.)
| | - Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (M.G.G.); (P.R.)
| | | | - Carlo Cifani
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.M.); (I.M.); (S.K.T.); (M.V.M.D.B.); (C.C.); (G.L.); (F.A.)
| | - Giulio Lupidi
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.M.); (I.M.); (S.K.T.); (M.V.M.D.B.); (C.C.); (G.L.); (F.A.)
| | - Francesco Amenta
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.M.); (I.M.); (S.K.T.); (M.V.M.D.B.); (C.C.); (G.L.); (F.A.)
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (M.G.G.); (P.R.)
| |
Collapse
|
48
|
Kayser B, Verges S. Hypoxia, energy balance, and obesity: An update. Obes Rev 2021; 22 Suppl 2:e13192. [PMID: 33470528 DOI: 10.1111/obr.13192] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
Because of the enduring rise in the prevalence of obesity worldwide, there is continued interest in hypoxia as a mechanism underlying the pathophysiology of obesity and its comorbidities and as a potential therapeutic adjunct for the management of the disease. Lifelong exposure to altitude is accompanied by a lower risk for obesity, whereas altitude sojourns are generally associated with a loss of body mass. A negative energy balance upon exposure to hypoxia can be due to a combination of changes in determinants of energy expenditure (resting metabolic rate and physical activity energy expenditure) and energy intake (appetite). Over the past 15 years, the potential therapeutic interest of hypobaric or normobaric hypoxic exposure in individuals with obesity-to lower body mass and improve health status-has become an active field of research. Various protocols have been implemented, using actual altitude sojourns or intermittent normobaric hypoxic exposures, at rest or in association with physical activity. Although several studies suggest benefits on body mass and cardiovascular and metabolic variables, further investigations are required before recommending hypoxic exposure in obesity management programs. Future studies should also better clarify the effects of hypoxia on appetite, the intestinal microbiota, and finally on overall energy balance.
Collapse
Affiliation(s)
- Bengt Kayser
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Samuel Verges
- HP2 Laboratory, INSERM, Grenoble Alpes University Hospital, Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
49
|
Francisqueti-Ferron FV, Garcia JL, Ferron AJT, Nakandakare-Maia ET, Gregolin CS, Silva JPDC, Dos Santos KC, Lo ÂTC, Siqueira JS, de Mattei L, de Paula BH, Sarzi F, Silva CCVDA, Moreto F, Costa MR, Ferreira ALA, Minatel IO, Corrêa CR. Gamma-oryzanol as a potential modulator of oxidative stress and inflammation via PPAR-y in adipose tissue: a hypothetical therapeutic for cytokine storm in COVID-19? Mol Cell Endocrinol 2021; 520:111095. [PMID: 33253762 PMCID: PMC7695949 DOI: 10.1016/j.mce.2020.111095] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/18/2022]
Abstract
The literature has reported a higher prevalence of negative clinical outcomes due to Coronavirus disease 19 (COVID-19) in obese individuals. This can be explained by the cytokine storm, result from the cytokine production from both obesity and viral infection. Gamma-oryzanol (γOz) is a compound with anti-inflammatory and antioxidant activities. However, little is known about the γOz action as a possible agonist of peroxisome proliferator-activated receptor gamma (PPAR-γ). The aim of this study was to test the hypothesis that γOz attenuates the cytokine storm by stimulating PPAR-γ in the adipose tissue. METHODS Male Wistar rats were randomly divided into three experimental groups and fed ad libitum for 30 weeks with control diet (C, n = 6), high sugar-fat diet (HSF, n = 6) or high sugar-fat diet + γOz (HSF + γOz, n = 6). HSF groups also received water + sucrose (25%). The γOz dose was 0.5% in the chow. Evaluation in animals included caloric intake, body weight, adiposity index, plasma triglycerides, and HOMA-IR. In adipose tissue was evaluated: PPAR-γ gene and protein expression, inflammatory and oxidative stress parameters, and histological analysis. RESULTS Adipose tissue dysfunction was observed in HSF group, which presented remarkable PPAR-γ underexpression and increased levels of cytokines, other inflammatory markers and oxidative stress. The γOz treatment prevented adipose tissue dysfunction and promoted PPAR-γ overexpression. CONCLUSION Natural compounds as γOz can be considered a coadjutant therapy to prevent the cytokine storm in COVID-19 patients with obesity conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Juliana Silva Siqueira
- São Paulo State University (Unesp), Institute of Bioscience, Botucatu, São Paulo, Brazil
| | - Letícia de Mattei
- São Paulo State University (Unesp), Medical School, Botucatu, São Paulo, Brazil
| | | | - Felipe Sarzi
- Botucatu Integrated College (UNIFAC), Botucatu, São Paulo, Brazil
| | | | - Fernando Moreto
- São Paulo State University (Unesp), Medical School, Botucatu, São Paulo, Brazil
| | | | | | - Igor Otávio Minatel
- São Paulo State University (Unesp), Institute of Bioscience, Botucatu, São Paulo, Brazil
| | | |
Collapse
|
50
|
Relationship between visceral adipose tissue and genetic mutations (VHL and KDM5C) in clear cell renal cell carcinoma. Radiol Med 2021; 126:645-651. [PMID: 33400184 DOI: 10.1007/s11547-020-01310-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/15/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The sequencing of the renal cell carcinoma (RCC) genome has detected several mutations with prognostic meaning. The association between visceral adipose tissue (VAT) and clear cell renal cell carcinoma (ccRCC) is well known. The relationship among abdominal adipose tissue distribution and ccRCC-VHL and KDM5C genetic mutations is, to the knowledge of the authors, not known. METHODS In this retrospective study, we enrolled 97 Caucasian male patients divided into three groups: the control group (n = 35), the ccRCC-VHL group (n = 52) composed of ccRCC patients with VHL mutations and ccRCC-KDM5C group (n = 10) composed of ccRCC patients with KDM5C mutation. Total adipose tissue (TAT) area, VAT area and subcutaneous adipose tissue (SAT) area were measured in the groups. VAT/SAT ratio was calculated for each subject. RESULTS Statistically significant differences between ccRCC-KDM5C group and ccRCC-VHL group were obtained for TAT area (p < 0.05), VAT area (p < 0.05) and VAT/SAT ratio (p < 0.05); between ccRCC-VHL group and control group for TAT area (p < 0.001) and VAT area (p < 0.01); and between ccRCC-KDM5C group and control group for TAT area (p < 0.0001), VAT area (p < 0.0001) and SAT area (p < 0.01). CONCLUSIONS This study demonstrates for the first time an increased amount of TAT, especially VAT, in the ccRCC-VHL and ccRCC-KDM5C groups. The effect was greater for the ccRCC-KDM5C group.
Collapse
|