1
|
Son YL, Hou J, Kato-Suzuki M, Okamatsu-Ogura Y, Watase M, Kiyonari H, Kondo T. Eva1 deficiency prevents obesity-induced metabolic disorders by reducing visceral adipose dysfunction. Metabolism 2025; 168:156235. [PMID: 40118448 DOI: 10.1016/j.metabol.2025.156235] [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: 01/17/2025] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
AIMS Epithelial V-like antigen 1 (Eva1) is a highly specific marker for brown adipose tissue (BAT) in both mice and humans, but its metabolic function remains unclear. We investigated the impact of Eva1 deletion on the development of obesity. METHODS To assess the metabolic role of Eva1, we generated whole-body and adipocyte-specific Eva1knockout (KO) mice, which were subjected to a high-fat diet (HFD) for 12 weeks and characterized metabolic phenotypes. To further elucidate the depot-dependent impact of Eva1 deficiency, we performed histological analysis and 3' mRNA-seq of BAT and epididymal visceral white adipose tissue (eWAT). To investigate the role of macrophage-derived Eva1 in obesity development, we transplanted wild-type (WT) or Eva1KO macrophages into Eva1KO mice fed an HFD. RESULTS We found that whole-body Eva1KO mice are resistant to HFD-induced obesity, insulin resistance and visceral adipose inflammation. However, Eva1 deletion in adipocytes, both brown and white, did not phenocopy these protective effects. Notably, whole-body Eva1 deficiency triggers functional changes in eWAT, but not in BAT. These results led us to investigate a possible involvement of macrophages in Eva1-mediated obesity regulation. We found that Eva1 is expressed in macrophages and plays a role in lipopolysaccharide (LPS)-induced inflammatory responses, possibly through the direct interaction with toll-like receptor 4 (TLR4). Moreover, Eva1KO mice exhibited improved survival rates in the face of severe sepsis induced by LPS. Importantly, transplantation of WT macrophages to Eva1KO mice abolished the beneficial effects of whole-body Eva1 deletion against obesity and visceral adipose inflammation. CONCLUSION Our findings highlight macrophage-derived Eva1 as an important mediator in obesity-induced eWAT remodeling, suggesting that targeting Eva1 could offer a novel therapeutic strategy for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- You Lee Son
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
| | - Jiahui Hou
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Mira Kato-Suzuki
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Megumi Watase
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Toru Kondo
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
2
|
Chung E, Zhang D, Gonzalez Porras M, Hsu CG. TREM2 as a regulator of obesity-induced cardiac remodeling: mechanisms and therapeutic insights. Am J Physiol Heart Circ Physiol 2025; 328:H1073-H1082. [PMID: 40152357 DOI: 10.1152/ajpheart.00075.2025] [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: 01/29/2025] [Revised: 02/17/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are global health challenges that significantly increase the risk of cardiovascular diseases (CVD). Advances in immunometabolism have identified triggering receptor expressed on myeloid cells 2 (TREM2) as a key regulator of macrophage function, lipid metabolism, and inflammation resolution. Although extensively studied in neurodegenerative diseases, TREM2's role in metabolic disorders and cardiovascular health is an emerging area of research. This review explores TREM2's molecular structure and functions, emphasizing its contributions to immunometabolic regulation in obesity and T2DM. Evidence from preclinical models demonstrates that TREM2 modulates macrophage-driven inflammatory responses, lipid clearance, plaque stability, fibrosis, and myocardial remodeling. Translational findings suggest that TREM2 expression correlates with cardiometabolic outcomes, underscoring its potential as a therapeutic target. Key knowledge gaps include TREM2's temporal dynamics during disease progression, sex-specific effects, and interactions with recruited or resident macrophage activation in obesity and T2DM. Integrating mechanistic and translational insights is critical to harness TREM2's immunoregulatory potential for improving CVD outcomes in metabolic disorders.
Collapse
Affiliation(s)
- Eunhee Chung
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, Texas, United States
| | - David Zhang
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States
| | - Maria Gonzalez Porras
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States
| | - Chia George Hsu
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, Texas, United States
| |
Collapse
|
3
|
Li Z, Cheng W, Gao K, Liang S, Ke L, Wang M, Fan J, Li D, Zhang P, Xu Z, Li N. Pyroptosis: A spoiler of peaceful coexistence between cells in degenerative bone and joint diseases. J Adv Res 2025; 71:227-262. [PMID: 38876191 DOI: 10.1016/j.jare.2024.06.010] [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: 02/17/2024] [Revised: 05/23/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND As people age, degenerative bone and joint diseases (DBJDs) become more prevalent. When middle-aged and elderly people are diagnosed with one or more disorders such as osteoporosis (OP), osteoarthritis (OA), and intervertebral disc degeneration (IVDD), it often signals the onset of prolonged pain and reduced functionality. Chronic inflammation has been identified as the underlying cause of various degenerative diseases, including DBJDs. Recently, excessive activation of pyroptosis, a form of programed cell death (PCD) mediated by inflammasomes, has emerged as a primary driver of harmful chronic inflammation. Consequently, pyroptosis has become a potential target for preventing and treating DBJDs. AIM OF REVIEW This review explored the physiological and pathological roles of the pyroptosis pathway in bone and joint development and its relation to DBJDs. Meanwhile, it elaborated the molecular mechanisms of pyroptosis within individual cell types in the bone marrow and joints, as well as the interplay among different cell types in the context of DBJDs. Furthermore, this review presented the latest compelling evidence supporting the idea of regulating the pyroptosis pathway for DBJDs treatment, and discussed the potential, limitations, and challenges of various therapeutic strategies involving pyroptosis regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW In summary, an interesting identity for the unregulated pyroptosis pathway in the context of DBJDs was proposed in this review, which was undertaken as a spoiler of peaceful coexistence between cells in a degenerative environment. Over the extended course of DBJDs, pyroptosis pathway perpetuated its activity through crosstalk among pyroptosis cascades in different cell types, thus exacerbating the inflammatory environment throughout the entire bone marrow and joint degeneration environment. Correspondingly, pyroptosis regulation therapy emerged as a promising option for clinical treatment of DBJDs.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jilin Fan
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000 China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300 China.
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
4
|
Tang P, Wang J, Tang X, Li Y, Li S. Insulin‑like growth factor 2 in spermatogenesis dysfunction (Review). Mol Med Rep 2025; 31:129. [PMID: 40116127 PMCID: PMC11938415 DOI: 10.3892/mmr.2025.13494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/21/2025] [Indexed: 03/23/2025] Open
Abstract
Spermatogenesis dysfunction is characterized by abnormal morphology, destruction, atrophy of seminiferous tubules, blocked differentiation of spermatogenic cells, decreased sperm count and increased sperm abnormalities. Inflammation, oxidative stress, endoplasmic reticulum stress and obesity are important factors leading to spermatogenesis dysfunction. It has been demonstrated that insulin‑like growth factor 2 (IGF2) is closely related to the aforementioned factors. In the present review, the relationship between IGF2 and inflammation, oxidative stress, ER stress and obesity was investigated, providing theoretical and experimental evidence on the role of IGF2 in the prevention and treatment of spermatogenesis dysfunction of male infertility.
Collapse
Affiliation(s)
- Pingping Tang
- Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jiale Wang
- Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiaohan Tang
- Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yichun Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital University of South China, Hengyang, Hunan 421001, P.R. China
| | - Suyun Li
- Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
5
|
Cerullo M, Armeli F, Mengoni B, Menin M, Crudeli ML, Businaro R. Curcumin Modulation of the Gut-Brain Axis for Neuroinflammation and Metabolic Disorders Prevention and Treatment. Nutrients 2025; 17:1430. [PMID: 40362738 PMCID: PMC12073396 DOI: 10.3390/nu17091430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Curcumin, a polyphenolic compound derived from Curcuma longa, has gained significant attention for its potential therapeutic benefits, particularly counteracting inflammation, oxidative stress, and metabolic disorders. Its chemical structure, featuring conjugated double bonds between two aromatic rings, allows it to act as an electron donor, thereby mitigating free radical formation. Despite its poor solubility in water, curcumin is stable in acidic environments and undergoes significant metabolism in both the liver and the gut. Intestinal microbiota, particularly at the colon level, further metabolizes curcumin into several derivatives, including dihydrocurcumin and tetrahydrocurcumin, which exhibit antioxidant and anti-inflammatory properties. Studies suggest that curcumin can reduce body mass index (BMI) and improve other body composition parameters, especially when used in combination with lifestyle changes, though its bioavailability is low due to its rapid metabolism and the resulting low blood concentration. In obesity, dysfunctional adipose tissue remodeling and chronic inflammation play critical roles in the development of metabolic complications. Curcumin's anti-inflammatory properties are related to the inhibition of the NF-κB pathway, leading to the reduction in inflammatory markers in adipocytes and macrophages. Additionally, curcumin modulates oxidative stress by activating the NRF2 pathway, enhancing cellular antioxidant defenses. Emerging evidence also supports curcumin's potential in improving gut health by modulating microbiota composition, enhancing intestinal barrier function, and reducing systemic inflammation. This interaction with the gut-brain axis highlights the broader implications of curcumin in neuroprotection, as it positively affects cognitive function and mitigates neuroinflammation in neurodegenerative diseases like Alzheimer's. disease. Thus, curcumin holds promise as a multifaceted agent in the management of obesity and associated diseases.
Collapse
Affiliation(s)
- Miriam Cerullo
- Neurofarba Department, University of Florence, 50139 Florence, Italy;
| | - Federica Armeli
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (F.A.); (B.M.); (M.M.); (M.L.C.)
| | - Beatrice Mengoni
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (F.A.); (B.M.); (M.M.); (M.L.C.)
| | - Martina Menin
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (F.A.); (B.M.); (M.M.); (M.L.C.)
| | - Maria Luisa Crudeli
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (F.A.); (B.M.); (M.M.); (M.L.C.)
| | - Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (F.A.); (B.M.); (M.M.); (M.L.C.)
| |
Collapse
|
6
|
Wu D, Eeda V, Maria Z, Rawal K, Wang A, Herlea-Pana O, Babu Undi R, Lim HY, Wang W. Targeting IRE1α improves insulin sensitivity and thermogenesis and suppresses metabolically active adipose tissue macrophages in male obese mice. eLife 2025; 13:RP100581. [PMID: 40244655 PMCID: PMC12005715 DOI: 10.7554/elife.100581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Overnutrition engenders the expansion of adipose tissue and the accumulation of immune cells, in particular, macrophages, in the adipose tissue, leading to chronic low-grade inflammation and insulin resistance. In obesity, several proinflammatory subpopulations of adipose tissue macrophages (ATMs) identified hitherto include the conventional 'M1-like' CD11C-expressing ATM and the newly discovered metabolically activated CD9-expressing ATM; however, the relationship among ATM subpopulations is unclear. The ER stress sensor inositol-requiring enzyme 1α (IRE1α) is activated in the adipocytes and immune cells under obesity. It is unknown whether targeting IRE1α is capable of reversing insulin resistance and obesity and modulating the metabolically activated ATMs. We report that pharmacological inhibition of IRE1α RNase significantly ameliorates insulin resistance and glucose intolerance in male mice with diet-induced obesity. IRE1α inhibition also increases thermogenesis and energy expenditure, and hence protects against high fat diet-induced obesity. Our study shows that the 'M1-like' CD11c+ ATMs are largely overlapping with but yet non-identical to CD9+ ATMs in obese white adipose tissue. Notably, IRE1α inhibition diminishes the accumulation of obesity-induced metabolically activated ATMs and 'M1-like' ATMs, resulting in the curtailment of adipose inflammation and ensuing reactivation of thermogenesis, without augmentation of the alternatively activated M2 macrophage population. Our findings suggest the potential of targeting IRE1α for the therapeutic treatment of insulin resistance and obesity.
Collapse
Affiliation(s)
- Dan Wu
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at BirminghamBirminghamUnited States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Venkateswararao Eeda
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Zahra Maria
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Komal Rawal
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | | | - Oana Herlea-Pana
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Ram Babu Undi
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Hui-Ying Lim
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at BirminghamBirminghamUnited States
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Weidong Wang
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at BirminghamBirminghamUnited States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| |
Collapse
|
7
|
Muszka Z, Jenei V, Mácsik R, Mezhonova E, Diyab S, Csősz R, Bácsi A, Mázló A, Koncz G. Life-threatening risk factors contribute to the development of diseases with the highest mortality through the induction of regulated necrotic cell death. Cell Death Dis 2025; 16:273. [PMID: 40216765 PMCID: PMC11992264 DOI: 10.1038/s41419-025-07563-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025]
Abstract
Chronic diseases affecting the cardiovascular system, diabetes mellitus, neurodegenerative diseases, and various other organ-specific conditions, involve different underlying pathological processes. However, they share common risk factors that contribute to the development and progression of these diseases, including air pollution, hypertension, obesity, high cholesterol levels, smoking and alcoholism. In this review, we aim to explore the connection between four types of diseases with different etiologies and various risk factors. We highlight that the presence of risk factors induces regulated necrotic cell death, leading to the release of damage-associated molecular patterns (DAMPs), ultimately resulting in sterile inflammation. Therefore, DAMP-mediated inflammation may be the link explaining how risk factors can lead to the development and maintenance of chronic diseases. To explore these processes, we summarize the main cell death pathways activated by the most common life-threatening risk factors, the types of released DAMPs and how these events are associated with the pathophysiology of diseases with the highest mortality. Various risk factors, such as smoking, air pollution, alcoholism, hypertension, obesity, and high cholesterol levels induce regulated necrosis. Subsequently, the release of DAMPs leads to chronic inflammation, which increases the risk of many diseases, including those with the highest mortality rates.
Collapse
Affiliation(s)
- Zsuzsa Muszka
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Viktória Jenei
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Rebeka Mácsik
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Evgeniya Mezhonova
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Silina Diyab
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Réka Csősz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Anett Mázló
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| | - Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| |
Collapse
|
8
|
Mujkić R, Šnajder Mujkić D, Rožac K, Matić A, Kovač Lukić T, Divković D, Selthofer-Relatić K. Converted Macrophage Polarization and Expression of COL6α3-Early Predictors of Remodeling Processes in Adipose Tissue of Male Children. Biomedicines 2025; 13:935. [PMID: 40299533 PMCID: PMC12025005 DOI: 10.3390/biomedicines13040935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Overweight and obesity in early childhood is a serious public health problem as in most cases it persists into adulthood and significantly affects the quality of life. The aim of this study was to investigate the mechanisms that trigger extracellular matrix (ECM) remodeling in the subcutaneous (SAT) and visceral (VAT) adipose tissue of male children in relation to their body weight. Methods: During elective abdominal surgery, SAT and VAT were acquired from 75 male subjects undergoing hernia repair (inguinal herniorrhaphy by Ferguson) or orchidopexy. Based on their Z-score, subjects were separated into two groups. The morphometry of both adipose tissue compartments was assessed after hematoxylin and eosin histological staining, immunohistochemistry to quantify CD163+ cells and the number of crown-like structures (CLSs), and real-time polymerase chain reaction to assess the relative gene expression for collagen VI subtype alpha 3 (COL6α3). Results: Obese and overweight individuals were found to have higher numbers of CD163+ cells, greater numbers of CLSs in VAT and SAT, and a higher expression of COL6α3 in both compartments. Conclusions: Obesity in childhood may lead to increased COL6α3 gene expression and promote the activation of macrophage polarization, compromise the structural integrity of the ECM, and thus influence the development of inflammatory processes.
Collapse
Affiliation(s)
- Robert Mujkić
- Department of Anatomy, Histology, Embryology, Pathological Anatomy and Pathological Histology, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia; (K.R.); (T.K.L.)
| | - Darija Šnajder Mujkić
- Department of Anatomy and Neuroscience, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia;
- Clinical Institute of Nuclear Medicine and Radiation Protection, University Hospital Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - Karla Rožac
- Department of Anatomy, Histology, Embryology, Pathological Anatomy and Pathological Histology, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia; (K.R.); (T.K.L.)
| | - Anita Matić
- Department of Pathophysiology, Physiology and Immunology, Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia;
| | - Tanja Kovač Lukić
- Department of Anatomy, Histology, Embryology, Pathological Anatomy and Pathological Histology, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia; (K.R.); (T.K.L.)
| | - Dalibor Divković
- Department of Pediatric Surgery, University Hospital Osijek, J. Huttlera 4, 31000 Osijek, Croatia;
- Department of Surgery, Urology, Orthopedics and Physical and Rehabilitation Medicine, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - Kristina Selthofer-Relatić
- Department of Cardiovascular Disease, University Hospital Osijek, J. Huttlera 4, 31000 Osijek, Croatia;
- Department of Pathophysiology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, 3100 Osijek, Croatia
| |
Collapse
|
9
|
Sun Y, Shan X, Li M, Niu Y, Sun Z, Ma X, Wang T, Zhang J, Niu D. Autoimmune mechanisms and inflammation in obesity-associated type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease. Funct Integr Genomics 2025; 25:84. [PMID: 40205260 DOI: 10.1007/s10142-025-01587-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025]
Abstract
Obesity, characterized by the excessive accumulation of white adipose tissue, is a significant global health burden and a major risk factor for a range of diseases, including malignancies and metabolic disorders. Individuals with high visceral fat content are particularly susceptible to severe complications such as type 2 diabetes, cardiovascular diseases, and liver disorders. However, the pathogenesis of obesity-related metabolic diseases extends beyond simple adiposity. Chronic obesity triggers a prolonged inflammatory response, which leads to tissue fibrosis and sustained organ damage, contributing to multi-organ dysfunction. This review explores the autoimmune mechanisms and inflammatory pathways underlying obesity-induced type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease, with an emphasis on their interrelated pathophysiology and the potential for therapeutic interventions.
Collapse
Grants
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
Collapse
Affiliation(s)
- Yuanyuan Sun
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Xueting Shan
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Mingyang Li
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Yifan Niu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Zhongxin Sun
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Tao Wang
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, 211300, Jiangsu, China.
| | - Jufang Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China.
| |
Collapse
|
10
|
Zhou H, Gizlenci M, Xiao Y, Martin F, Nakamori K, Zicari EM, Sato Y, Tullius SG. Obesity-associated Inflammation and Alloimmunity. Transplantation 2025; 109:588-596. [PMID: 39192462 PMCID: PMC11868468 DOI: 10.1097/tp.0000000000005183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Obesity is a worldwide health problem with a rapidly rising incidence. In organ transplantation, increasing numbers of patients with obesity accumulate on waiting lists and undergo surgery. Obesity is in general conceptualized as a chronic inflammatory disease, potentially impacting alloimmune response and graft function. Here, we summarize our current understanding of cellular and molecular mechanisms that control obesity-associated adipose tissue inflammation and provide insights into mechanisms affecting transplant outcomes, emphasizing on the beneficial effects of weight loss on alloimmune responses.
Collapse
Affiliation(s)
- Hao Zhou
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Merih Gizlenci
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany
| | - Yao Xiao
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Friederike Martin
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of Surgery, CVK/CCM, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Keita Nakamori
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Elizabeth M. Zicari
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Faculté de Pharmacie, Université Paris Cité, Paris, France
| | - Yuko Sato
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Stefan G. Tullius
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
11
|
Michelini S, Greco S, Vaia N, Puleo V, Pellegrino P, Di Vincenzo A, Michelini S, Herbst KL, Goteri G, Luca T, Castorina S, Giordano A, Ciarmela P, Cinti S. Endothelial cell alterations in capillaries of adipose tissue from patients affected by lipedema. Obesity (Silver Spring) 2025; 33:695-708. [PMID: 40077894 PMCID: PMC11937865 DOI: 10.1002/oby.24244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/28/2024] [Accepted: 12/19/2024] [Indexed: 03/14/2025]
Abstract
OBJECTIVE This study aimed to evaluate adipose tissue of lipedema patients. METHODS Gluteo-femoral (affected area) and interscapular (nonaffected area) adipose tissue from 10 lean patients affected by lipedema stage 1 to 2 was studied and compared with tissue from 10 patients with obesity and 12 lean patients. RESULTS The main features were alterations of capillaries with wall thickening (p ≤ 0.0001), endothelial and pericyte hyperplasia (p = 0.03 and p = 0.004), hypodense areas in basal membrane, and endothelial degeneration with exfoliation of degenerated cells into the capillary lumen. Adipocytes were larger (hypertrophic) in affected (P ≤ 0.0001) and nonaffected (p = 0.0003) areas compared with those with obesity and who were lean (both p ≤ 0.0001). Frequently the cytoplasm of adipocytes contained massive deposition of calcium crystals as revealed by Von Kossa staining (p = 0.023) and electron microscopy. CD68 immunoreactive macrophages were more abundant in affected areas (p = 0.005), and their number was similar to that found in fat from patients with obesity (p = 0.17). Despite adipocyte hypertrophy and inflammation, lack of the healthy marker perilipin-1 and the presence of crown-like structures were only rarely seen, while they were quite frequent in patients with obesity. CONCLUSIONS Our data support the idea that cell alterations happen in the early stages of adipocyte development (endothelium/pericyte) in the adipose organ of women affected by lipedema.
Collapse
Affiliation(s)
- Sandro Michelini
- Vascular Diagnostics and Rehabilitation ServiceMarino HospitalRomeItaly
- Physical Medicine and RehabilitationSan Giovanni Battista HospitalRomeItaly
| | - Stefania Greco
- Department of Experimental and Clinical Medicine, Center of ObesityUniversità Politecnica delle Marche (Polytechnic University of Marche)AnconaItaly
| | - Nicola Vaia
- Plastic, Reconstructive and Aesthetic SurgeryEuropean HospitalRomeItaly
| | - Valeria Puleo
- Department of Science and Public HealthCatholic University Policlinico GemelliRomeItaly
| | - Pamela Pellegrino
- Department of Experimental and Clinical Medicine, Center of ObesityUniversità Politecnica delle Marche (Polytechnic University of Marche)AnconaItaly
| | - Angelica Di Vincenzo
- Department of Experimental and Clinical Medicine, Center of ObesityUniversità Politecnica delle Marche (Polytechnic University of Marche)AnconaItaly
| | - Serena Michelini
- Physical Medicine and RehabilitationSan Giovanni Battista HospitalRomeItaly
| | | | - Gaia Goteri
- Department of Biomedical Sciences and Public Health, Section of Pathological Anatomy and HistopathologyUniversità Politecnica delle Marche (Polytechnic University of Marche)AnconaItaly
| | - Tonia Luca
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia"University of CataniaCataniaItaly
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia"University of CataniaCataniaItaly
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Center of ObesityUniversità Politecnica delle Marche (Polytechnic University of Marche)AnconaItaly
- Istituto di Ricovero e Cura a Carattere Scientifico (Scientific Institute for Research, Hospitalization and Health Care), Istituto Nazionale di Ricovero e Cura per Anziani (National Institute of Hospitalization and Care for the Elderly) (IRCCS/INRCA)AnconaItaly
| | - Pasquapina Ciarmela
- Department of Experimental and Clinical Medicine, Center of ObesityUniversità Politecnica delle Marche (Polytechnic University of Marche)AnconaItaly
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of ObesityUniversità Politecnica delle Marche (Polytechnic University of Marche)AnconaItaly
| |
Collapse
|
12
|
Bianchi F, Roccabianca P, Vianello E, Gentile G, La Sala L, Bandera F, Tacchini L, Zoia R, Corsi Romanelli MM, Dozio E. Inhibition of DPP-4 Attenuates Endotoxemia-Induced NLRC4 Inflammasome and Inflammation in Visceral Adipose Tissue of Mice Fed a High-Fat Diet. Biomolecules 2025; 15:333. [PMID: 40149869 PMCID: PMC11940500 DOI: 10.3390/biom15030333] [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: 01/22/2025] [Revised: 02/12/2025] [Accepted: 02/23/2025] [Indexed: 03/29/2025] Open
Abstract
Inflammasomes are protein complexes that trigger pro-inflammatory responses and promote many diseases, including adipose tissue dysfunction. Linagliptin (L), a DPP-4 inhibitor used for type 2 diabetes therapy, has putative anti-inflammatory effects. This work explores L effects on inflammasome regulation, inflammation, and adipose tissue dysfunction in obese mice. Male C57BL/6N mice were fed a normal chow (NC) diet, high-fat (HF) diet, or HF diet with L (HFL) for 15 weeks. Gene expression and histological examinations were performed on visceral (VAT) and subcutaneous (SAT) adipose tissue samples. Biomarkers were quantified on sera. Murine macrophages were utilized for in vitro analyses. L decreased HF-induced endotoxemia and circulating inflammatory indicators. Despite having no effect on body weight, L reduced VAT inflammation by decreasing endotoxemia-induced NLRC4 inflammasome, inflammation severity, and fat cell hypertrophy. Although SAT response differed from VAT, inflammation was slightly reduced in this tissue too. In vitro, L modulated inflammation by directly reducing the pro-inflammatory macrophage phenotype. In obesity, increased NLRC4 inflammasome expression links endotoxemia and VAT inflammation. L protected against endotoxemia, maybe by affecting gut permeability and VAT responses. The decreased polarization of macrophages toward a pro-inflammatory phenotype and the reduction in adipocyte hypertrophy are involved in the response to L.
Collapse
Affiliation(s)
- Francesca Bianchi
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
| | - Paola Roccabianca
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, 26900 Lodi, Italy;
| | - Elena Vianello
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- Laboratorio Sperimentale Ricerche Biomarcatori di Danno d’Organo, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Guendalina Gentile
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
| | - Lucia La Sala
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- IRCCS MultiMedica, 20138 Milan, Italy
| | - Francesco Bandera
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- IRCCS MultiMedica, 20138 Milan, Italy
| | - Lorenza Tacchini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- Laboratorio Sperimentale Ricerche Biomarcatori di Danno d’Organo, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Riccardo Zoia
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
| | - Massimiliano M. Corsi Romanelli
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- Dipartimento di Patologia Clinica e Sperimentale, IRCCS Istituto Auxologico, 20149 Milan, Italy
| | - Elena Dozio
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- Laboratorio Sperimentale Ricerche Biomarcatori di Danno d’Organo, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| |
Collapse
|
13
|
Scheidl TB, Wager JL, Thompson JA. Adipose Tissue Stromal Cells: Rheostats for Adipose Tissue Function and Metabolic Disease Risk. Can J Cardiol 2025:S0828-282X(25)00137-0. [PMID: 39986382 DOI: 10.1016/j.cjca.2025.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025] Open
Abstract
The transition from metabolically healthy obesity to the development of obesity-associated metabolic syndrome and cardiovascular disease is thought to be triggered by a loss in the functional integrity of adipose tissue. Although mature adipocytes are the primary functional units that carry out lipid partitioning in adipose tissue for the promotion of whole-body energy balance, they are supported by a heterogenous collection of nonadipocytes in the stroma. Research over the past couple of decades has expanded perspectives on the homeostatic and pathological roles of the nonadipocyte compartment. Adipose progenitors originate in the embryonic period and drive the developmental adipogenesis that establishes the set point of adiposity. A population of adipocyte progenitors reside in adult depots and serve an important homeostatic role as a reservoir to support adipocyte turnover. Adipocyte hypertrophy in obesity increases the rate of adipocyte death and the ability of progenitors to support this high rate of adipocyte turnover is important for the preservation of the lipid-buffering function of adipose tissue. Some evidence exists to suggest that impaired adipogenesis or a decline in progenitors capable of differentiation is a key event in the development of adipose dysfunction. The efficiency of macrophages to clear the debris and toxic lipids released from dead adipocytes lies at the fulcrum between preservation of adipose function and the progression toward chronic inflammation. Although macrophages in collaboration with other immune cells propagate the inflammation that underlies adipose dysfunction, there is now a greater appreciation for the diverse and unique roles of immune cells within adipose tissue.
Collapse
Affiliation(s)
- Taylor B Scheidl
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada. https://twitter.com/TaylorScheidl
| | - Jessica L Wager
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jennifer A Thompson
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
14
|
Luévano-Martínez LA, Méndez-Fernández A, Rueda-Munguía M, Bonilla-Ruelas D, Brunck MEG, García-Rivas G. Glycerol metabolism is activated in both palmitic acid-stimulated and adipose tissue macrophages from a murine model of cardiometabolic heart failure. J Physiol 2025. [PMID: 39977539 DOI: 10.1113/jp287791] [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: 10/09/2024] [Accepted: 01/29/2025] [Indexed: 02/22/2025] Open
Abstract
Macrophages chronically exposed to saturated fatty acids, such as those encountered in adipose tissue, present a pro-inflammatory phenotype with a characteristic foamy morphology. This feature is caused by the excess uptake of circulating lipids, yielding large cytoplasmic lipid bodies formed by triacylglycerols and cholesteryl derivatives. Palmitic acid (PA) is a potent inflammatory inducer in macrophages after chronic exposure to this fatty acid. However, acute exposure to this fatty acid is unable to activate a pro-inflammatory phenotype, although it is sufficient to induce metabolic reprogramming including the formation of small lipid bodies. In the present study, we used an in vitro model of human monocyte-derived macrophages to unravel the early stages of metabolic reprogramming observed in macrophages exposed to PA. We observed that partial inhibition of the glycerol 3-phosphate shuttle is necessary for supplying glycerol 3-phosphate for triacylglycerol biosynthesis. Furthermore, we characterized an alternative pathway to increase the concentration of glycerol 3-phosphate involving an aquaporin and glycerol kinase. Our results suggested that early lipid bodies biogenesis rises as a response mechanism to buffer excessive PA without inducing a pro-inflammatory program. Additionally, we observed that macrophages chronically exposed to PA eventually upregulate the production of inflammatory cytokines. Finally, our in vitro observations were confirmed in adipose tissue macrophages derived from a preclinical mouse model of cardiometabolic heart failure with preserved ejection fraction characterized by heightened adiposity and inflammation. KEY POINTS: The glycerol 3-phosphate shuttle is partially inhibited in palmitic acid-activated macrophages. Aquaporin 3 expression is upregulated in macrophages exposed to palmitic acid and in adipose tissue macrophages from a murine model of cardiometabolic heart failure. Aquaporin 3 participates in the biosynthesis of triacylglycerols by supplying extracellular glycerol.
Collapse
Affiliation(s)
- Luis Alberto Luévano-Martínez
- Tecnologico de Monterrey, Centro de Investigación Biomédica, Hospital Zambrano Hellion, TecSalud, San Pedro Garza García, Nuevo León, México
- The Institute for Obesity Research, Tecnologico de Monterrey, Tecnologico, Monterrey, Nuevo León, Mexico
| | - Abraham Méndez-Fernández
- Tecnologico de Monterrey, Centro de Investigación Biomédica, Hospital Zambrano Hellion, TecSalud, San Pedro Garza García, Nuevo León, México
| | - Mayte Rueda-Munguía
- Tecnologico de Monterrey, Centro de Investigación Biomédica, Hospital Zambrano Hellion, TecSalud, San Pedro Garza García, Nuevo León, México
| | - Diana Bonilla-Ruelas
- The Institute for Obesity Research, Tecnologico de Monterrey, Tecnologico, Monterrey, Nuevo León, Mexico
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Nuevo León, Mexico
| | - Marion E G Brunck
- The Institute for Obesity Research, Tecnologico de Monterrey, Tecnologico, Monterrey, Nuevo León, Mexico
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Nuevo León, Mexico
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Centro de Investigación Biomédica, Hospital Zambrano Hellion, TecSalud, San Pedro Garza García, Nuevo León, México
- The Institute for Obesity Research, Tecnologico de Monterrey, Tecnologico, Monterrey, Nuevo León, Mexico
| |
Collapse
|
15
|
Peng C, Jiang H, Jing L, Yang W, Guan X, Wang H, Yu S, Cao Y, Wang M, Ma H, Lv Z, Gu H, Xia C, Guo X, Sun B, Wang A, Xie C, Wu W, Lu L, Song J, Lei S, Wu R, Zang Y, Tang E, Li J. Macrophage SUCLA2 coupled glutaminolysis manipulates obesity through AMPK. Nat Commun 2025; 16:1738. [PMID: 39966410 PMCID: PMC11836283 DOI: 10.1038/s41467-025-57044-w] [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: 02/10/2024] [Accepted: 02/09/2025] [Indexed: 02/20/2025] Open
Abstract
Obesity is regarded as a chronic inflammatory disease involving adipose tissue macrophages (ATM), but whether immunometabolic reprogramming of ATM affects obesity remains unclarified. Here we show that in ATM glutaminolysis is the fundamental metabolic flux providing energy and substrate, bridging with AMP-activated protein kinase (AMPK) activity, succinate-induced interleukin-1β (IL-1β) production, and obesity. Abrogation of AMPKα in myeloid cells promotes proinflammatory ATM, impairs thermogenesis and energy expenditure, and aggravates obesity in mice fed with high-fat diet (HFD). Conversely, IL-1β neutralization or myeloid IL-1β abrogation prevents obesity caused by AMPKα deficiency. Mechanistically, ATP generated from glutaminolysis suppresses AMPK to decrease phosphorylation of the β subunit of succinyl-CoA synthetase (SUCLA2), thereby resulting in the activation of succinyl-CoA synthetase and the overproduction of succinate and IL-1β; by contrast, siRNA-mediated SUCLA2 knockdown reduces obesity induced by HFD in mice. Lastly, phosphorylated SUCLA2 in ATM correlates negatively with obesity in humans. Our results thus implicate a glutaminolysis/AMPK/SUCLA2/IL-1β axis of inflammation and obesity regulation in ATM.
Collapse
Grants
- 32000525 National Natural Science Foundation of China (National Science Foundation of China)
- 82273983 National Natural Science Foundation of China (National Science Foundation of China)
- National Key R&D Program of China (2022YFA1303800),Science and Technology Commission of Shanghai Municipality (23ZR1474700),Shanghai Institute of Materia Medica, Chinese Academy of Science (New Star project)
- China Postdoctoral Science Foundation (2024M760704)
- the Research Funds of Hangzhou Institute for Advanced Study, UCAS (2024HIAS-N001)
- the Research Funds of Hangzhou Institute for Advanced Study, UCAS (No. 2022ZZ01013, and. 2023HIAS-V002)
- Medical Innovation Research Special Project of Shanghai (19411971500), Medical Innovation Research Special Project of Shanghai (22Y11908600), Science and Technology Commission of Yangpu District (YPM202101 and YPGWM202401)
- the National Natural Science Foundation of China grants (82130099, and 92253306),the Research Funds of Hangzhou Institute for Advanced Study, UCAS (2023HIAS-Y030)
Collapse
Affiliation(s)
- Chang Peng
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Haowen Jiang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Liya Jing
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Lingang Laboratory, Shanghai, 201203, China
| | - Wenhua Yang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China
| | - Xiaotong Guan
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Hanlin Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Sike Yu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Yutang Cao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Min Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Huan Ma
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Zan Lv
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- College of Pharmacy, Fudan University, Shanghai, 210023, China
| | - Hongyu Gu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China
| | - Chunmei Xia
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Bin Sun
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China
- Institute of Gastrointestinal Surgery and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China
| | - Aili Wang
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China
- Institute of Gastrointestinal Surgery and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wenbiao Wu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Luyiyi Lu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Jiayi Song
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Saifei Lei
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Rui Wu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Yi Zang
- Lingang Laboratory, Shanghai, 201203, China
| | - Erjiang Tang
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China.
- Institute of Gastrointestinal Surgery and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China.
| | - Jia Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
16
|
Lee MJ. Vitamin D Enhancement of Adipose Biology: Implications on Obesity-Associated Cardiometabolic Diseases. Nutrients 2025; 17:586. [PMID: 39940444 PMCID: PMC11820181 DOI: 10.3390/nu17030586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Vitamin D is activated into 1α,25(OH)2D through two hydroxylation steps that are primarily catalyzed by 25-hydroxylase in the liver and 1α-hydroxylase in the kidneys. The active form of vitamin D regulates myriads of cellular functions through its nuclear receptor, vitamin D receptor (VDR). Vitamin D metabolizing enzymes and VDR are expressed in adipose tissues and vitamin D regulates multiple aspects of adipose biology including the recruitment and differentiation of adipose stem cells into adipocytes and metabolic, endocrine, and immune properties. Obesity is associated with low vitamin D status, which is thought to be explained by its sequestration in large mass of adipose tissues as well as dysregulated vitamin D metabolism. Low vitamin D status in obesity may negatively impact adipose biology leading to adipose tissue dysfunctions, the major pathological factors for cardiometabolic diseases in obesity. In this review, the current understanding of vitamin D metabolism and its molecular mechanisms of actions, focusing on vitamin D-VDR regulation of adipose biology with their implications on obesity-associated diseases, is discussed. Whether improving vitamin D status leads to reductions in adiposity and risks for cardiometabolic diseases is also discussed.
Collapse
Affiliation(s)
- Mi-Jeong Lee
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| |
Collapse
|
17
|
Kumar GVN, Wang RS, Sharma AX, David NL, Amorim T, Sinden DS, Doshi NK, Wabitsch M, Gingras S, Ejaz A, Rubin JP, Maron BA, Fazeli PK, Steinhauser ML. Non-canonical lysosomal lipolysis drives mobilization of adipose tissue energy stores with fasting. Nat Commun 2025; 16:1330. [PMID: 39900947 PMCID: PMC11790841 DOI: 10.1038/s41467-025-56613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Physiological adaptations to fasting enable humans to survive for prolonged periods without food and involve molecular pathways that may drive life-prolonging effects of dietary restriction in model organisms. Mobilization of fatty acids and glycerol from adipocyte lipid stores by canonical neutral lipases, including the rate limiting adipose triglyceride lipase (Pnpla2/ATGL), is critical to the adaptive fasting response. Here we discovered an alternative mechanism of lipolysis in adipocytes involving a lysosomal program. We functionally tested lysosomal lipolysis with pharmacological and genetic approaches in mice and in murine and human adipocyte and adipose tissue explant culture, establishing dependency on lysosomal acid lipase (LIPA/LAL) and the microphthalmia/transcription factor E (MiT/TFE) family. Our study establishes a model whereby the canonical pathway is critical for rapid lipolytic responses to adrenergic stimuli operative in the acute stage of fasting, while the alternative lysosomal pathway dominates with prolonged fasting.
Collapse
Affiliation(s)
- G V Naveen Kumar
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rui-Sheng Wang
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ankit X Sharma
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Natalie L David
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tânia Amorim
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Daniel S Sinden
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nandini K Doshi
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Martin Wabitsch
- University Medical Center Department of Pediatrics and Adolescent Medicine, Ulm, Germany
| | - Sebastien Gingras
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - J Peter Rubin
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bradley A Maron
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- The University of Maryland-Institute for Health Computing, Bethesda, MD, USA
| | - Pouneh K Fazeli
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew L Steinhauser
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Huang L, Zhang T, Zhu Y, Lai X, Tao H, Xing Y, Li Z. Deciphering the Role of CD36 in Gestational Diabetes Mellitus: Linking Fatty Acid Metabolism and Inflammation in Disease Pathogenesis. J Inflamm Res 2025; 18:1575-1588. [PMID: 39925938 PMCID: PMC11806725 DOI: 10.2147/jir.s502314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/27/2025] [Indexed: 02/11/2025] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common pregnancy complications which exerts detrimental effects on mothers and children. Emerging evidence has pointed to the important role of the fatty acid transporter protein CD36 in the pathogenesis of GDM. As a heavily glycosylated transmembrane protein, CD36 is widely expressed in diverse cell types, including placental trophoblasts, monocytes/macrophages, adipocytes, and pancreatic cells et al. CD36 plays a key role in lipid metabolism and signal transduction in the pathophysiological mechanism of GDM. The modified expression and functionality of CD36 may contribute to inflammation and oxidative stress in maternal tissues, interfere with insulin signaling, and subsequently influence maternal insulin sensitivity and fetal growth, increasing the risk for GDM. This review provides an overview of the current knowledge regarding the expression and function of CD36 in various tissues throughout pregnancy and explores how CD36 dysregulation can activate inflammatory pathways, worsen insulin resistance, and disrupt lipid metabolism, thereby complicating the necessary metabolic adjustments during pregnancy. Furthermore, the review delves into emerging therapeutic approaches targeting CD36 signaling to alleviate the impacts of GDM. Understanding the involvement of CD36 in GDM could yield crucial insights into its mechanisms and potential interventions for enhancing maternal and fetal health outcomes.
Collapse
Affiliation(s)
- Li Huang
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, Sichuan, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, People’s Republic of China
| | - Tong Zhang
- Department of Laboratory Medicine, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yuanyuan Zhu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Xueling Lai
- Shenzhen Guangming Maternal & Child Healthcare Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Hualin Tao
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, Sichuan, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, People’s Republic of China
| | - Yuhan Xing
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Zhaoyinqian Li
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, Sichuan, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
19
|
Alaamri S, Serafi AS, Hussain Z, Bafail SK, Bafail MA, Demirkhanyan L, Gondi CS, Sohail S. Overweight-Related Hypertension in Middle-Aged Men Is Linked to Elevated Leptin, TNF-α, IL-6, Cholesterol, and Reduced Testosterone. PATHOPHYSIOLOGY 2025; 32:7. [PMID: 39982363 PMCID: PMC11843874 DOI: 10.3390/pathophysiology32010007] [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: 11/05/2024] [Revised: 01/04/2025] [Accepted: 01/27/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND/OBJECTIVES One of the major causes of hypertension (HT) is the transition of normal weight (NW) status to overweight (OW) status and obesity in a population, which leads to cardiovascular disease (CVD) and other disorders. A variety of factors/variables are involved in the development of HT and OW-related hypertension (OHT). However, we planned to investigate the pathophysiological role of serum leptin (Lep), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), total cholesterol (TC) and serum testosterone (ST) in OHT in middle-aged men. METHODS We consulted three groups of middle-aged men (age: 51-60 years)-an HT group (n: 97, high normal weight (HNW), body mass index (BMI): 23-24.9 kg/m2); an OHT group (n: 97, high overweight (HOW), BMI: 28-29.9 kg/m2) and a normal control group (NC, n: 98, HNW)-to investigate the variations in and correlations of Lep, IL-6, TNF-α, ST, TC and other variables. RESULTS Significant variations were obtained for the comparisons of TNF-α, Lep, ST and TC for the patient groups. OHT vs. NC showed a significant difference for ST. OHT vs. NC and OHT vs. HT had significant variations for IL-6. Significant changes were obtained for the serum levels of TNF-α, Lep, IL-6, ST and TC among groups. Significant and positive linear associations were obtained for TNF-α, Lep, TC and IL-6. Significant and negative linear associations were found for ST plotted against Lep, TNF-α and IL-6. CONCLUSIONS The current report provides pathophysiological evidence of the interactive role of serum Lep, TNF-α, ST, TC and IL-6 in middle-aged men with HT and OHT. We suggest that the changes we noted in the present study would be helpful for further BMI-based studies in various subcategories of NW, OW and obese subjects with/without HT.
Collapse
Affiliation(s)
- Shalan Alaamri
- Department of Medicine, College of Medicine, University of Jeddah, Jeddah 21589, Saudi Arabia;
| | - Abdulhalim S. Serafi
- Department of Physiology, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Zahir Hussain
- Department of Physiology, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Shouq K. Bafail
- Department of Biology, College of Sciences, Umm Al-Qura University, Aljamoum 22254, Saudi Arabia;
| | - Mohammed A. Bafail
- Department of Physiology, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Lusine Demirkhanyan
- Department of Internal Medicine, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (L.D.); (C.S.G.)
- Department of Surgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Christopher S. Gondi
- Department of Internal Medicine, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (L.D.); (C.S.G.)
- Department of Surgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Department of Health Science Education and Pathology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Sumera Sohail
- Department of Physiology, University of Karachi, Karachi 75270, Pakistan;
| |
Collapse
|
20
|
Di Porzio A, Barrella V, Cigliano L, Mauriello G, Troise AD, Scaloni A, Iossa S, Mazzoli A. Diet-induced impairment of skeletal muscle and adipose tissue metabolic homeostasis and its prevention by probiotic administration. Pflugers Arch 2025; 477:223-239. [PMID: 39537965 DOI: 10.1007/s00424-024-03041-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/27/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Western dietary pattern is one of the main contributors to the increased risk of obesity and chronic diseases, through oxidative stress and inflammation, that are the two key mechanisms targeting metabolic organs, such as skeletal muscle and adipose tissue. The chronic exposure to high levels of dietary fatty acids can increase the amount of intramyocellular lipids in skeletal muscle, altering glucose homeostasis and contributing to a reduction in mitochondrial oxidative capacity. Probiotic administration is a promising approach as preventive strategy to attenuate metabolic damage induced by Western diet. Here, we investigated the beneficial effect of Limosillactobacillus reuteri DSM 17938 on the inflammatory state and oxidative balance in the skeletal muscle and adipose tissue of adult rats fed a western diet for 8 weeks, focusing on the role of skeletal muscle mitochondria. Limosillactobacillus reuteri DSM 17938 administration protected the skeletal muscle from mitochondrial dysfunction and oxidative stress, preventing the establishment of inflammation and insulin resistance. Interestingly, a further beneficial effect of the probiotic was exerted on body composition, favoring the deposition of protein mass and preventing adipose tissue hypertrophy and inflammation. These results open the possibility for the use of this probiotic in therapeutic approaches for nutrition-related diseases.
Collapse
Affiliation(s)
- Angela Di Porzio
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
| | - Valentina Barrella
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
- National Biodiversity Future Center, 90133, Palermo, Italy
| | - Luisa Cigliano
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Gianluigi Mauriello
- Department of Agricultural Sciences, University of Naples Federico II, 80055, Portici, Italy
| | - Antonio Dario Troise
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, Institute for the Animal Production System in the Mediterranean Environment, National Research Council, 80055, Portici, Italy
| | - Andrea Scaloni
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, Institute for the Animal Production System in the Mediterranean Environment, National Research Council, 80055, Portici, Italy
| | - Susanna Iossa
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy.
- National Biodiversity Future Center, 90133, Palermo, Italy.
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy.
| | - Arianna Mazzoli
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy.
| |
Collapse
|
21
|
Gálvez I, Navarro M, Torres-Piles S, Martín-Cordero L, Hinchado M, Ortega E. Exercise-induced anxiety impairs local and systemic inflammatory response and glucose metabolism in C57BL/6J mice. Brain Behav Immun Health 2025; 43:100910. [PMID: 39686921 PMCID: PMC11648794 DOI: 10.1016/j.bbih.2024.100910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction The complex physiological and psychological responses to regular exercise are yet to be fully elucidated. Exercise strongly modulates the immune system, inducing a plethora of dynamic responses involving the innate immune cell function and inflammatory processes that contribute to both potential health benefits and harmful side effects. Indeed, the relationship between physical exercise, stress, immunity, and metabolism serves as a paramount model of neuroimmunoendocrine interaction. Thus, the objective of this study was to conduct a comprehensive analysis of both systemic and local immunophysiological responses together with behavioral responses to a protocol of anxiety-inducing exercise. Material and methods C57BL/6J mice were randomly allocated into sedentary or exercised groups, where the anxiety-inducing exercise protocol was based on a 14-day consecutive program of swimming in water at 38 °C. Anxiety-like behavior was corroborated through the elevated plus maze test. Systemic biomarkers of the stress response were assessed using ELISA technique and the expression of systemic inflammatory cytokines with Bio-Plex system. Phagocytic/microbicide activity, the expression of M1/M2 phenotype markers (CD11c, iNOS, CD206, ARG-1) and cytokines of the inflammatory response (MCP-1, IL-8, IL-6, TNF-α, TGF-β, IL-10) of peritoneal macrophages were determined via flow cytometry. Adipose tissue macrophage infiltration was studied through fluorescence immunohistochemistry. Results Anxiety-like behavior, elevated circulating glucose concentrations, systemic stress and inflammatory responses, together with increased oxidative stress and inflammatory profile of peritoneal macrophages, and macrophage infiltration in white adipose tissue were observed in exercised animals. Conclusions A protocol of exercise that induces anxiety is associated with a neuroimmunoendocrine dysregulation affecting the feedback between the inflammatory and the stress responses, together with detrimental metabolic effects in glucose modulation. Systemic inflammatory alterations are accompanied by detrimental inflammatory responses in tissue macrophage populations. Altogether, these results show that exercise associated with anxiety, stress, pro-inflammatory responses, and hyperglycaemia represents a model of 'dangerous exercise'.
Collapse
Affiliation(s)
- I. Gálvez
- Immunophysiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071, Badajoz, Spain
- Immunophysiology Research Group, Nursing Department, Faculty of Medicine and Health Sciences, University of Extremadura, 06071, Badajoz, Spain
| | - M.C. Navarro
- Immunophysiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071, Badajoz, Spain
- Immunophysiology Research Group, Physiology Department, Faculty of Sciences, University of Extremadura, 06071, Badajoz, Spain
| | - S. Torres-Piles
- Immunophysiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071, Badajoz, Spain
- Immunophysiology Research Group, Department of Medical-Surgical Therapy, Faculty of Medicine, University of Extremadura, 06071, Badajoz, Spain
| | - L. Martín-Cordero
- Immunophysiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071, Badajoz, Spain
- Immunophysiology Research Group, Nursing Department, University Center of Plasencia, University of Extremadura, 10600, Plasencia, Spain
| | - M.D. Hinchado
- Immunophysiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071, Badajoz, Spain
- Immunophysiology Research Group, Physiology Department, Faculty of Sciences, University of Extremadura, 06071, Badajoz, Spain
| | - E. Ortega
- Immunophysiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071, Badajoz, Spain
- Immunophysiology Research Group, Physiology Department, Faculty of Sciences, University of Extremadura, 06071, Badajoz, Spain
| |
Collapse
|
22
|
Han SM, Nahmgoong H, Yim KM, Kim JB. How obesity affects adipocyte turnover. Trends Endocrinol Metab 2025; 36:147-160. [PMID: 39095230 DOI: 10.1016/j.tem.2024.07.004] [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: 04/18/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024]
Abstract
Cellular turnover is fundamental for tissue homeostasis and integrity. Adipocyte turnover, accounting for 4% of the total cellular mass turnover in humans, is essential for adipose tissue homeostasis during metabolic stress. In obesity, an altered adipose tissue microenvironment promotes adipocyte death. To clear dead adipocytes, macrophages are recruited and form a distinctive structure known as crown-like structure; subsequently, new adipocytes are generated from adipose stem and progenitor cells in the adipogenic niche to replace dead adipocytes. Accumulating evidence indicates that adipocyte death, clearance, and adipogenesis are sophisticatedly orchestrated during adipocyte turnover. In this Review, we summarize our current understandings of each step in adipocyte turnover, discussing its key players and regulatory mechanisms.
Collapse
Affiliation(s)
- Sang Mun Han
- National Leader Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hahn Nahmgoong
- National Leader Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung Min Yim
- National Leader Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae Bum Kim
- National Leader Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
23
|
Wu D, Eeda V, Maria Z, Rawal K, Wang A, Herlea-Pana O, Undi RB, Lim HY, Wang W. Targeting IRE1α improves insulin sensitivity and thermogenesis and suppresses metabolically active adipose tissue macrophages in male obese mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.17.603931. [PMID: 39071288 PMCID: PMC11275733 DOI: 10.1101/2024.07.17.603931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Overnutrition engenders the expansion of adipose tissue and the accumulation of immune cells, in particular, macrophages, in the adipose tissue, leading to chronic low-grade inflammation and insulin resistance. In obesity, several proinflammatory subpopulations of adipose tissue macrophages (ATMs) identified hitherto include the conventional "M1-like" CD11C-expressing ATM and the newly discovered metabolically activated CD9-expressing ATM; however, the relationship among ATM subpopulations is unclear. The ER stress sensor inositol-requiring enzyme 1α (IRE1α) is activated in the adipocytes and immune cells under obesity. It is unknown whether targeting IRE1α is capable of reversing insulin resistance and obesity and modulating the metabolically activated ATMs. We report that pharmacological inhibition of IRE1α RNase significantly ameliorates insulin resistance and glucose intolerance in male mice with diet-induced obesity. IRE1α inhibition also increases thermogenesis and energy expenditure, and hence protects against high fat diet-induced obesity. Our study shows that the "M1-like" CD11c+ ATMs are largely overlapping with but yet non-identical to CD9+ ATMs in obese white adipose tissue. Notably, IRE1α inhibition diminishes the accumulation of obesity-induced metabolically activated ATMs and "M1-like" ATMs, resulting in the curtailment of adipose inflammation and ensuing reactivation of thermogenesis, without augmentation of the alternatively activated M2 macrophage population. Our findings suggest the potential of targeting IRE1α for the therapeutic treatment of insulin resistance and obesity.
Collapse
Affiliation(s)
- Dan Wu
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, Alabama, 35233, United States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Venkateswararao Eeda
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Zahra Maria
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Komal Rawal
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Audrey Wang
- Indian Springs School, 190 Woodward Dr, Pelham, Alabama 35124
| | - Oana Herlea-Pana
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Ram Babu Undi
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Hui-Ying Lim
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, Alabama, 35233, United States
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Weidong Wang
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, Alabama, 35233, United States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| |
Collapse
|
24
|
Wang T, Zhou D, Hong Z. Sarcopenia and cachexia: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2025; 6:e70030. [PMID: 39764565 PMCID: PMC11702502 DOI: 10.1002/mco2.70030] [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: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/17/2025] Open
Abstract
Sarcopenia is defined as a muscle-wasting syndrome that occurs with accelerated aging, while cachexia is a severe wasting syndrome associated with conditions such as cancer and immunodeficiency disorders, which cannot be fully addressed through conventional nutritional supplementation. Sarcopenia can be considered a component of cachexia, with the bidirectional interplay between adipose tissue and skeletal muscle potentially serving as a molecular mechanism for both conditions. However, the underlying mechanisms differ. Recognizing the interplay and distinctions between these disorders is essential for advancing both basic and translational research in this area, enhancing diagnostic accuracy and ultimately achieving effective therapeutic solutions for affected patients. This review discusses the muscle microenvironment's changes contributing to these conditions, recent therapeutic approaches like lifestyle modifications, small molecules, and nutritional interventions, and emerging strategies such as gene editing, stem cell therapy, and gut microbiome modulation. We also address the challenges and opportunities of multimodal interventions, aiming to provide insights into the pathogenesis and molecular mechanisms of sarcopenia and cachexia, ultimately aiding in innovative strategy development and improved treatments.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Zhen Hong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| |
Collapse
|
25
|
Tye H, Conos SA, Djajawi TM, Gottschalk TA, Abdoulkader N, Kong IY, Kammoun HL, Narayana VK, Kratina T, Speir M, Emery J, Simpson DS, Hall C, Vince AJ, Russo S, Crawley R, Rashidi M, Hildebrand JM, Murphy JM, Whitehead L, De Souza DP, Masters SL, Samson AL, Lalaoui N, Hawkins ED, Murphy AJ, Vince JE, Lawlor KE. Divergent roles of RIPK3 and MLKL in high-fat diet-induced obesity and MAFLD in mice. Life Sci Alliance 2025; 8:e202302446. [PMID: 39532538 PMCID: PMC11557689 DOI: 10.26508/lsa.202302446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Cell death frequently occurs in the pathogenesis of obesity and metabolic dysfunction-associated fatty liver disease (MAFLD). However, the exact contribution of core cell death machinery to disease manifestations remains ill-defined. Here, we show via the direct comparison of mice genetically deficient in the essential necroptotic regulators, receptor-interacting protein kinase-3 (RIPK3) and mixed lineage kinase domain-like (MLKL), as well as mice lacking apoptotic caspase-8 in myeloid cells combined with RIPK3 loss, that RIPK3/caspase-8 signaling regulates macrophage inflammatory responses and drives adipose tissue inflammation and MAFLD upon high-fat diet feeding. In contrast, MLKL, divergent to RIPK3, contributes to both obesity and MAFLD in a manner largely independent of inflammation. We also uncover that MLKL regulates the expression of molecules involved in lipid uptake, transport, and metabolism, and congruent with this, we discover a shift in the hepatic lipidome upon MLKL deletion. Collectively, these findings highlight MLKL as an attractive therapeutic target to combat the growing obesity pandemic and metabolic disease.
Collapse
Affiliation(s)
- Hazel Tye
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
| | - Stephanie A Conos
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Tirta M Djajawi
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Timothy A Gottschalk
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Nasteho Abdoulkader
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
| | - Isabella Y Kong
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | | | - Vinod K Narayana
- Metabolomics Australia, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Australia
| | | | - Mary Speir
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Jack Emery
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Daniel S Simpson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Cathrine Hall
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Angelina J Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Sophia Russo
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Rhiannan Crawley
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Maryam Rashidi
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Joanne M Hildebrand
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - James M Murphy
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - David P De Souza
- Metabolomics Australia, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Australia
| | - Seth L Masters
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Andre L Samson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Najoua Lalaoui
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Edwin D Hawkins
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | | | - James E Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Kate E Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| |
Collapse
|
26
|
Wang Z, Liu X, Sheng L, Xie Y, Feng W, Yu L. Effects of duration of high-fat diet on adipocyte hyperplasia in rat epididymis. Obes Res Clin Pract 2025; 19:54-62. [PMID: 39922761 DOI: 10.1016/j.orcp.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 06/25/2024] [Accepted: 02/01/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND High-fat diet (HFD) contributes to obesity and enhances the expression of mature adipocyte markers. However, the effect of HFD on adipocyte hyperplasia remains controversial. This may be due to variations in the duration of HFD. This study aimed to investigate the effects of different durations of HFD on adipocyte hyperplasia and the expression of mature adipocyte-related markers in obese rats. METHODS We divided 32 Sprague-Dawley rats into four groups: B (standard diet control), H1 (HFD for four weeks), H2 (HFD for eight weeks), and H3 (HFD for 12 weeks). We evaluated the morphological changes in epididymal fat cells, measured serum inflammatory markers using enzyme-linked immunosorbent assay (ELISA) kits, and quantified adipocyte hyperplasia and maturation markers using western blotting. RESULTS We observed progressive increases in body weight, epididymal fat weight, serum leptin, TNF-α, IL-6, irisin, PPARγ, adiponectin, and FNDC5 protein expression over 8 weeks of HFD. 12 weeks of HFD intervention resulted in significant decreases in irisin, PPARγ, adiponectin, and FNDC5. Concurrently, the expression of perilipin A and ATGL declined with prolonged HFD. CONCLUSIONS Our results suggest that the duration of HFD significantly affects adipocyte ability to undergo hyperplasia in the epididymis of obese rats. Specifically, 4 weeks of HFD did not change the capacity for adipocyte hyperplasia, while 8 weeks of the diet enhanced this capacity. Interestingly, a longer diet duration (12 weeks) led to a decrease in adipocyte hyperplasia.
Collapse
Affiliation(s)
- Zhaoxin Wang
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Xiujuan Liu
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China.
| | - Lei Sheng
- Department of Scientific Research, Nanjing Sport Institute, Nanjing, China.
| | - Yuting Xie
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Wanyu Feng
- Department of Science Experiment Center, Nanjing Sport Institute, Nanjing, China
| | - Li Yu
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| |
Collapse
|
27
|
Diao B, Fan Z, Zhou B, Zhan H. Crosstalk between pancreatic cancer and adipose tissue: Molecular mechanisms and therapeutic implications. Biochem Biophys Res Commun 2024; 740:151012. [PMID: 39561650 DOI: 10.1016/j.bbrc.2024.151012] [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: 08/03/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
The incidence rate of pancreatic cancer, a fatal illness with a meager 5-year survival rate, has been on the rise in recent times. When individuals accumulate excessive amounts of adipose tissue, the adipose organ becomes dysfunctional due to alterations in the adipose tissue microenvironment associated with inflammation and metabolism. This phenomenon may potentially contribute to the aberrant accumulation of fat that initiates pancreatic carcinogenesis, thereby influencing the disease's progression, resistance to treatment, and metastasis. This review presents a summary of the impact of pancreatic steatosis, visceral fat, cancer-associated adipocytes and lipid diets on the advancement of pancreatic cancer, as well as the reciprocal effects of pancreatic cancer on adipose tissue. Understanding the molecular mechanisms underlying the relationship between dysfunctional adipose tissue and pancreatic cancer better may lead to the discovery of new therapeutic targets for the disease's prevention and individualized treatment. This is especially important given the rising global incidence of obesity, which will improve the pancreatic cancer treatment options that are currently insufficient.
Collapse
Affiliation(s)
- Boyu Diao
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
28
|
Evans AJ, Li YL. Remodeling of the Intracardiac Ganglia During the Development of Cardiovascular Autonomic Dysfunction in Type 2 Diabetes: Molecular Mechanisms and Therapeutics. Int J Mol Sci 2024; 25:12464. [PMID: 39596529 PMCID: PMC11594459 DOI: 10.3390/ijms252212464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the most significant health issues worldwide, with associated healthcare costs estimated to surpass USD 1054 billion by 2045. The leading cause of death in T2DM patients is the development of cardiovascular disease (CVD). In the early stages of T2DM, patients develop cardiovascular autonomic dysfunction due to the withdrawal of cardiac parasympathetic activity. Diminished cardiac parasympathetic tone can lead to cardiac arrhythmia-related sudden cardiac death, which accounts for 50% of CVD-related deaths in T2DM patients. Regulation of cardiovascular parasympathetic activity is integrated by neural circuitry at multiple levels including afferent, central, and efferent components. Efferent control of cardiac parasympathetic autonomic tone is mediated through the activity of preganglionic parasympathetic neurons located in the cardiac extensions of the vagus nerve that signals to postganglionic parasympathetic neurons located in the intracardiac ganglia (ICG) on the heart. Postganglionic parasympathetic neurons exert local control on the heart, independent of higher brain centers, through the release of neurotransmitters, such as acetylcholine. Structural and functional alterations in cardiac parasympathetic postganglionic neurons contribute to the withdrawal of cardiac parasympathetic tone, resulting in arrhythmogenesis and sudden cardiac death. This review provides an overview of the remodeling of parasympathetic postganglionic neurons in the ICG, and potential mechanisms contributing to the withdrawal of cardiac parasympathetic tone, ventricular arrhythmogenesis, and sudden cardiac death in T2DM. Improving cardiac parasympathetic tone could be a therapeutic avenue to reduce malignant ventricular arrhythmia and sudden cardiac death, increasing both the lifespan and improving quality of life of T2DM patients.
Collapse
Affiliation(s)
- Anthony J. Evans
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
29
|
Miracle CE, McCallister CL, Egleton RD, Salisbury TB. Mechanisms by which obesity regulates inflammation and anti-tumor immunity in cancer. Biochem Biophys Res Commun 2024; 733:150437. [PMID: 39074412 PMCID: PMC11455618 DOI: 10.1016/j.bbrc.2024.150437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/12/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024]
Abstract
Obesity is associated with an increased risk for 13 different cancers. The increased risk for cancer in obesity is mediated by obesity-associated changes in the immune system. Obesity has distinct effects on different types of inflammation that are tied to tumorigenesis. For example, obesity promotes chronic inflammation in adipose tissue that is tumor-promoting in peripheral tissues. Conversely, obesity inhibits acute inflammation that rejects tumors. Obesity therefore promotes cancer by differentially regulating chronic versus acute inflammation. Given that obesity is chronic, the initial inflammation in adipose tissue will lead to systemic inflammation that could induce compensatory anti-inflammatory reactions in peripheral tissues to suppress chronic inflammation. The overall effect of obesity in peripheral tissues is therefore dependent on the duration and severity of obesity. Adipose tissue is a complex tissue that is composed of many cell types in addition to adipocytes. Further, adipose tissue cellularity is different at different anatomical sites throughout the body. Consequently, the sensitivity of adipose tissue to obesity is dependent on the anatomical location of the adipose depot. For example, obesity induces more inflammation in visceral than subcutaneous adipose tissue. Based on these studies, the mechanisms by which obesity promotes tumorigenesis are multifactorial and immune cell type-specific. The objective of our paper is to discuss the cellular mechanisms by which obesity promotes tumorigenesis by regulating distinct types of inflammation in adipose tissue and the tumor microenvironment.
Collapse
Affiliation(s)
- Cora E Miracle
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Chelsea L McCallister
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Richard D Egleton
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Travis B Salisbury
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| |
Collapse
|
30
|
Zhu BT, Liao QQ, Tian HY, Yu DJ, Xie T, Sun XL, Zhou XM, Han YX, Zhao YJ, El-Kassas M, Liu XX, Sun XD, Zhang YY. Estrogen: the forgotten player in metaflammation. Front Pharmacol 2024; 15:1478819. [PMID: 39575382 PMCID: PMC11578702 DOI: 10.3389/fphar.2024.1478819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
Metaflammation is low-grade inflammation triggered by chronic metabolic imbalance and caused by dysregulated metabolites in metabolic inflammatory syndrome (MIS), which includes four diseases: obesity, type 2 diabetes mellitus (T2DM), atherosclerosis (AS), and nonalcoholic fatty liver diseases (NAFLD, recently proposed to be replaced by metabolic dysfunction-associated steatotic liver disease, MASLD). These diseases exhibit apparent sex dimorphism as regards MIS. Estrogen not only plays a crucial role in gender differences in adults but also possesses an anti-inflammatory effect on many metabolic diseases. In this study, we present a prediction of the differential proteins and signal transduction of estrogen in MIS through network pharmacology and review the validated studies on obesity, T2DM, AS, and NAFLD. Subsequently, we compared them to obtain valuable targets, identify current gaps, and provide perspectives for future research on the mechanisms of estrogen in metaflammation.
Collapse
Affiliation(s)
- Bao-Ting Zhu
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Qing-Qing Liao
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Hai-Ying Tian
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Dao-Jiang Yu
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Teng Xie
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xi-Lu Sun
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xin-Meng Zhou
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ying-Xuan Han
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yu-Jie Zhao
- Medical College, Tibet University, Lasa, China
| | - Mohamed El-Kassas
- Endemic Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt
- Liver Disease Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Steatotic Liver Disease Study Foundation in Middle East and North Africa (SLMENA), Cairo, Egypt
| | - Xiu-Xiu Liu
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xiao-Dong Sun
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Medical College, Tibet University, Lasa, China
| | - Yuan-Yuan Zhang
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| |
Collapse
|
31
|
Xiao Q, Tang L, Chen S, Mei Y, Wang C, Yang J, Shang J, Li S, Wang W. Two-Pronged Attack: Dual Activation of Fat Reduction Using Near-Infrared-Responsive Nanosandwich for Targeted Anti-Obesity Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406985. [PMID: 39324577 DOI: 10.1002/advs.202406985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/06/2024] [Indexed: 09/27/2024]
Abstract
Excessive fat accumulation and chronic inflammation are two typical characteristics of obesity. AMP-activated protein kinase (AMPK), a master regulator of energy metabolism, is involved in adipogenesis, lipogenesis, and inflammation modulation in adipose tissue (AT). Thus, effective lipid reduction and anti-inflammation through AMPK regulation play vital roles in treating obesity. Herein, an anti-obesity nanosandwich is fabricated through attaching polymetformin (PolyMet) onto photothermal agent black phosphorus nanosheets (BP). PolyMet activates AMPK to inhibit adipogenesis, promote browning, and mitigate AT inflammation by decreasing macrophage infiltration, repolarizing macrophage phenotype, and downregulating pro-inflammatory cytokines. Additionally, BP induces lipolysis and apoptosis of adipocytes and macrophages through a photothermal effect. By further functionalization using hyaluronic acid (HA) and MMP2 substrate-linking P3 peptide-modified HA (P3-HA), an enhanced anti-obesity effect is obtained by dual-targeting of P3 and HA, and HA-mediated CD44 poly-clustering after MMP2 cleavage. Upon laser irradiation, the designed nanosandwich (P3-HA/PM@BP) effectively inhibits obesity development in obese mice, increases M2/M1 ratio in AT, reduces the serum levels of cholesterol/triglyceride and improves insulin sensitivity, exhibiting promising research potential to facilitate the clinical development of modern anti-obesity therapies.
Collapse
Affiliation(s)
- Qiaqia Xiao
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Lu Tang
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Siying Chen
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Yijun Mei
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Chuying Wang
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Jing Yang
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Jing Shang
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Shengliang Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| |
Collapse
|
32
|
Wolf M, Brochhausen C, Ramakrishnan V, Iberl S, Roth J, Seitz S, Burkhardt R, Stadler SC. Histologic Characterization of Tumor-Adjacent Mammary Adipose Tissue in Normal-Weight and Overweight/Obese Patients with Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:3515. [PMID: 39456610 PMCID: PMC11506523 DOI: 10.3390/cancers16203515] [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: 09/03/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Obesity is a risk factor of several types of cancer, including breast cancer. In this study, we aimed to histologically characterize the adipose tissue of the tumor microenvironment (TME) of triple-negative breast cancer (TNBC) in overweight/obese versus normal-weight patients. Methods: TNBC tissue sections from normal-weight (BMI<25) and overweight/obese patients (BMI≥25) were stained with antibodies against CD68, CD163, CD31, CD34, and vimentin. At the invasive tumor front, positive cells were counted in tumor adjacent adipose tissue (AT) and within cancer tissue (CT). Further, the size of the tumor-adjacent and distant mammary adipocytes was determined in perilipin stained sections. Expression of ANGPTL4, CD36 and FABP4, proteins involved in fatty acid metabolism, was analyzed in marginal tumor cells using an immune reactive score. Results: Overweight/obese TNBC patients had significantly larger adipocytes, higher numbers of CD163+ macrophages (BMI<25: 2.80 vs. BMI≥25: 10.45; p = 0.011) and lower numbers of CD31+ (BMI<25: 4.20 vs. BMI≥25: 2.40; p = 0.018) and CD34+ (BMI<25: 14.60 vs. BMI≥25: 5.20; p = 0.045) cells as markers of angiogenesis in the AT as well as a higher frequency of cancer-associated-fibroblast-like cells in the AT and CT (BMI<25: 7.60 vs. BMI≥25: 25.39 in total; p = 0.001). Moreover, expression of CD36 (BMI<25: 2.15 vs. BMI≥25: 2.60; p = 0.041) and ANGPTL4 (BMI<25: 6.00 vs. BMI≥25: 9.80; p = 0.026) was elevated in the TNBC cells of overweight/obese patients. Conclusions: Our data suggest BMI-related changes in the TME of overweight/obese TNBC patients, including hypertrophied adipocytes, reduced vascularization, more M2-like macrophages and CAF-like cells, and an increase in the expression of fatty acid metabolizing proteins in marginal tumor cells, all contributing to a more tumor-promoting, immunosuppressive environment.
Collapse
Affiliation(s)
- Marietta Wolf
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany
| | - Christoph Brochhausen
- Institute of Pathology, Medical Faculty Mannheim, University Heidelberg, 69120 Mannheim, Germany
- Institute of Pathology, Regensburg University, 93053 Regensburg, Germany
| | | | - Sabine Iberl
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
| | - Jonas Roth
- Department of Gynecology and Obstetrics, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Stephan Seitz
- Department of Gynecology and Obstetrics, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
| | - Sonja C. Stadler
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
| |
Collapse
|
33
|
Ali G, Zeb A, Usman M, Al‐Babili S. Walnut extract protects against hepatic inflammation and toxicity induced by a high-fat diet. Food Sci Nutr 2024; 12:8340-8352. [PMID: 39479714 PMCID: PMC11521631 DOI: 10.1002/fsn3.4405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 11/02/2024] Open
Abstract
A high-fat diet (HFD) is one of the main causes of obesity and metabolic diseases. The liver is particularly affected by HFD causing metabolic dysfunction associated with fatty liver disease. Therefore, different strategies are used to mitigate the negative effects of HFD. This study aimed to assess the protective effects of walnut extract against HFD-induced toxicity in mice. The mice were fed HFD and walnut extract alone or in combination. The walnut extract was analyzed for composition using high-performance liquid chromatography with a diode array detector (HPLC-DAD) and ultra-high-performance liquid chromatography with mass spectrometry (UHPLC-MS/MS). Serum lipid profile; liver histology; hepatic antioxidants such as catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), lipid peroxidation (TBARS), and reduced glutathione (GSH); inflammatory markers like IL-6 and TNF-α; and phospholipids were determined. Results showed that phenolic acids, epicatechin, catechin, benzaldehyde, and juglone were the main constituents in the extract. The HFD group showed increased hepatic fat accumulation as evidenced by biochemical and histopathological examinations compared to the control animals. The HFD group mice also showed increased body and cardiac weights, modified lipid profiles, decreased antioxidant status, and increased levels of hepatic inflammatory markers. The weights of the body and heart, lipid profiles, antioxidant contents (CAT, SOD, GSH-Px, TBARS, and GSH), and pro-inflammatory cytokines (IL-6 and TNF-α) were all normalized by consuming walnut extract. Similarly, the HFD group had significantly high amounts of hepatic lipase, phospholipid, and lysophospholipid levels, which were improved by walnut extract. In conclusion, walnut extract has been shown to play a unique role in promoting the recovery of liver damage caused by a high-fat diet.
Collapse
Affiliation(s)
- Gauhar Ali
- Department of BiotechnologyUniversity of MalakandChakdaraPakistan
| | - Alam Zeb
- Bioactive Lab, Centre of Excellence for Sustainable Food SecurityKing Abdullah University of Science and TechnologyThuwalKingdom of Saudi Arabia
- Department of BiochemistryUniversity of MalakandChakdaraPakistan
| | - Muhammad Usman
- Department of Basic SciencesUniversity of Veterinary and Animals SciencesNarowalPakistan
| | - Salim Al‐Babili
- Bioactive Lab, Centre of Excellence for Sustainable Food SecurityKing Abdullah University of Science and TechnologyThuwalKingdom of Saudi Arabia
| |
Collapse
|
34
|
Luo JH, Wang FX, Zhao JW, Yang CL, Rong SJ, Lu WY, Chen QJ, Zhou Q, Xiao J, Wang YN, Luo X, Li Y, Song DN, Chen C, Zhang CL, Chen SH, Yang P, Xiong F, Yu QL, Zhang S, Liu SW, Sun F, Wang CY. PDIA3 defines a novel subset of adipose macrophages to exacerbate the development of obesity and metabolic disorders. Cell Metab 2024; 36:2262-2280.e5. [PMID: 39293433 DOI: 10.1016/j.cmet.2024.08.009] [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: 11/02/2023] [Revised: 06/04/2024] [Accepted: 08/26/2024] [Indexed: 09/20/2024]
Abstract
Adipose tissue macrophages (ATMs) play important roles in maintaining adipose tissue homeostasis and orchestrating metabolic inflammation. Given the extensive functional heterogeneity and phenotypic plasticity of ATMs, identification of the authentically pathogenic ATM subpopulation under obese setting is thus necessitated. Herein, we performed single-nucleus RNA sequencing (snRNA-seq) and unraveled a unique maladaptive ATM subpopulation defined as ATF4hiPDIA3hiACSL4hiCCL2hi inflammatory and metabolically activated macrophages (iMAMs), in which PDIA3 is required for the maintenance of their migratory and pro-inflammatory properties. Mechanistically, ATF4 serves as a metabolic stress sensor to transcribe PDIA3, which then imposes a redox control on RhoA activity and strengthens the pro-inflammatory and migratory properties of iMAMs through RhoA-YAP signaling. Administration of Pdia3 small interfering RNA (siRNA)-loaded liposomes effectively repressed adipose inflammation and high-fat diet (HFD)-induced obesity. Together, our data support that strategies aimed at targeting iMAMs by suppressing PDIA3 expression or activity could be a viable approach against obesity and metabolic disorders in clinical settings.
Collapse
Affiliation(s)
- Jia-Hui Luo
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fa-Xi Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jia-Wei Zhao
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun-Liang Yang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan-Jie Rong
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wan-Ying Lu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi-Jie Chen
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Zhou
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Xiao
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Nan Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xi Luo
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Li
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan-Ni Song
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cai Chen
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng-Liang Zhang
- Department of Pharmacy, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Su-Hua Chen
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xiong
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi-Lin Yu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi-Wei Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, The Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Taiyuan, China
| | - Fei Sun
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Cong-Yi Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, The Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Taiyuan, China; The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Tongji Hospital Research Building, Wuhan, China.
| |
Collapse
|
35
|
Ishihara T, Kato K, Matsumoto K, Tanaka M, Hara A, Shiraki Y, Morisaki H, Urano Y, Ando R, Ito K, Mii S, Esaki N, Furuhashi K, Takefuji M, Suganami T, Murohara T, Enomoto A. Meflin/ISLR is a marker of adipose stem and progenitor cells in mice and humans that suppresses white adipose tissue remodeling and fibrosis. Genes Cells 2024; 29:902-920. [PMID: 39136356 PMCID: PMC11555626 DOI: 10.1111/gtc.13154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 10/04/2024]
Abstract
Identifying specific markers of adipose stem and progenitor cells (ASPCs) in vivo is crucial for understanding the biology of white adipose tissues (WAT). PDGFRα-positive perivascular stromal cells represent the best candidates for ASPCs. This cell lineage differentiates into myofibroblasts that contribute to the impairment of WAT function. However, ASPC marker protein(s) that are functionally crucial for maintaining WAT homeostasis are unknown. We previously identified Meflin as a marker of mesenchymal stem cells (MSCs) in bone marrow and tissue-resident perivascular fibroblasts in various tissues. We also demonstrated that Meflin maintains the undifferentiated status of MSCs/fibroblasts. Here, we show that Meflin is expressed in WAT ASPCs. A lineage-tracing experiment showed that Meflin+ ASPCs proliferate in the WAT of obese mice induced by a high-fat diet (HFD), while some of them differentiate into myofibroblasts or mature adipocytes. Meflin knockout mice fed an HFD exhibited a significant fibrotic response as well as increases in adipocyte cell size and the number of crown-like structures in WAT, accompanied by impaired glucose tolerance. These data suggested that Meflin expressed by ASPCs may have a role in reducing disease progression associated with WAT dysfunction.
Collapse
Grants
- 22ck0106779h0001 Japan Agency for Medical Research and Development
- 23gm1210009s0105 Japan Agency for Medical Research and Development
- 20H03467 Ministry of Education, Culture, Sports, Science and Technology
- 22H02848 Ministry of Education, Culture, Sports, Science and Technology
- 22K18390 Ministry of Education, Culture, Sports, Science and Technology
- Japan Agency for Medical Research and Development
- Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Toshikazu Ishihara
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Katsuhiro Kato
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Kotaro Matsumoto
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Miyako Tanaka
- Department of ImmunometabolismNagoya University Graduate School of MedicineNagoyaJapan
- Department of Molecular Medicine and MetabolismResearch Institute of Environmental Medicine, Nagoya UniversityNagoyaJapan
| | - Akitoshi Hara
- Center for Cardiovascular Research, University of Hawaii at ManoaHonoluluHawaiiUSA
| | - Yukihiro Shiraki
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Hidenori Morisaki
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yuya Urano
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Ryota Ando
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Kisuke Ito
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Shinji Mii
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Nobutoshi Esaki
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Kazuhiro Furuhashi
- Department of NephrologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Mikito Takefuji
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Takayoshi Suganami
- Department of ImmunometabolismNagoya University Graduate School of MedicineNagoyaJapan
- Department of Molecular Medicine and MetabolismResearch Institute of Environmental Medicine, Nagoya UniversityNagoyaJapan
| | - Toyoaki Murohara
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Atsushi Enomoto
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced StudyGifuJapan
| |
Collapse
|
36
|
Soedono S, Julietta V, Nawaz H, Cho KW. Dynamic Roles and Expanding Diversity of Adipose Tissue Macrophages in Obesity. J Obes Metab Syndr 2024; 33:193-212. [PMID: 39324219 PMCID: PMC11443328 DOI: 10.7570/jomes24030] [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] [Received: 09/14/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024] Open
Abstract
Adipose tissue macrophages (ATMs) are key regulators of adipose tissue (AT) inflammation and insulin resistance in obesity, and the traditional M1/M2 characterization of ATMs is inadequate for capturing their diversity in obese conditions. Single-cell transcriptomic profiling has revealed heterogeneity among ATMs that goes beyond the old paradigm and identified new subsets with unique functions. Furthermore, explorations of their developmental origins suggest that multiple differentiation pathways contribute to ATM variety. These advances raise concerns about how to define ATM functions, how they are regulated, and how they orchestrate changes in AT. This review provides an overview of the current understanding of ATMs and their updated categorization in both mice and humans during obesity. Additionally, diverse ATM functions and contributions in the context of obesity are discussed. Finally, potential strategies for targeting ATM functions as therapeutic interventions for obesity-induced metabolic diseases are addressed.
Collapse
Affiliation(s)
- Shindy Soedono
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Vivi Julietta
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Hadia Nawaz
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Kae Won Cho
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| |
Collapse
|
37
|
Sinha SK, Carpio MB, Nicholas SB. Fiery Connections: Macrophage-Mediated Inflammation, the Journey from Obesity to Type 2 Diabetes Mellitus and Diabetic Kidney Disease. Biomedicines 2024; 12:2209. [PMID: 39457523 PMCID: PMC11503991 DOI: 10.3390/biomedicines12102209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
The high prevalence of diabetes mellitus (DM) poses a significant public health challenge, with diabetic kidney disease (DKD) as one of its most serious consequences. It has become increasingly clear that type 2 DM (T2D) and the complications of DKD are not purely metabolic disorders. This review outlines emerging evidence related to the step-by-step contribution of macrophages to the development and progression of DKD in individuals who specifically develop T2D as a result of obesity. The macrophage is a prominent inflammatory cell that contributes to obesity, where adipocyte hypertrophy leads to macrophage recruitment and eventually to the expansion of adipose tissue. The recruited macrophages secrete proinflammatory cytokines, which cause systemic inflammation, glucose dysregulation, and insulin sensitivity, ultimately contributing to the development of T2D. Under such pathological changes, the kidney is susceptible to elevated glucose and thereby activates signaling pathways that ultimately drive monocyte recruitment. In particular, the early recruitment of proinflammatory macrophages in the diabetic kidney produces inflammatory cytokines/chemokines that contribute to inflammation and tissue damage associated with DKD pathology. Macrophage activation and recruitment are crucial inciting factors that also persist as DKD progresses. Thus, targeting macrophage activation and function could be a promising therapeutic approach, potentially offering significant benefits for managing DKD at all stages of progression.
Collapse
Affiliation(s)
- Satyesh K. Sinha
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
- Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - Maria Beatriz Carpio
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| | - Susanne B. Nicholas
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| |
Collapse
|
38
|
Mirabelli M, Misiti R, Sicilia L, Brunetti FS, Chiefari E, Brunetti A, Foti DP. Hypoxia in Human Obesity: New Insights from Inflammation towards Insulin Resistance-A Narrative Review. Int J Mol Sci 2024; 25:9802. [PMID: 39337290 PMCID: PMC11432683 DOI: 10.3390/ijms25189802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Insulin resistance (IR), marked by reduced cellular responsiveness to insulin, and obesity, defined by the excessive accumulation of adipose tissue, are two intertwined conditions that significantly contribute to the global burden of cardiometabolic diseases. Adipose tissue, beyond merely storing triglycerides, acts as an active producer of biomolecules. In obesity, as adipose tissue undergoes hypertrophy, it becomes dysfunctional, altering the release of adipocyte-derived factors, known as adipokines. This dysfunction promotes low-grade chronic inflammation, exacerbates IR, and creates a hyperglycemic, proatherogenic, and prothrombotic environment. However, the fundamental cause of these phenomena remains unclear. This narrative review points to hypoxia as a critical trigger for the molecular changes associated with fat accumulation, particularly within visceral adipose tissue (VAT). The activation of hypoxia-inducible factor-1 (HIF-1), a transcription factor that regulates homeostatic responses to low oxygen levels, initiates a series of molecular events in VAT, leading to the aberrant release of adipokines, many of which are still unexplored, and potentially affecting peripheral insulin sensitivity. Recent discoveries have highlighted the role of hypoxia and miRNA-128 in regulating the insulin receptor in visceral adipocytes, contributing to their dysfunctional behavior, including impaired glucose uptake. Understanding the complex interplay between adipose tissue hypoxia, dysfunction, inflammation, and IR in obesity is essential for developing innovative, targeted therapeutic strategies.
Collapse
Affiliation(s)
- Maria Mirabelli
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Roberta Misiti
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
- Operative Unit of Clinical Pathology, “Renato Dulbecco” Hospital, 88100 Catanzaro, Italy
| | - Luciana Sicilia
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Francesco S. Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
| | - Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Daniela P. Foti
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
- Operative Unit of Clinical Pathology, “Renato Dulbecco” Hospital, 88100 Catanzaro, Italy
| |
Collapse
|
39
|
Rosen ED, Kajimura S. Is it time to rethink the relationship between adipose inflammation and insulin resistance? J Clin Invest 2024; 134:e184663. [PMID: 39225103 PMCID: PMC11364379 DOI: 10.1172/jci184663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Affiliation(s)
- Evan D. Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
40
|
Singhal AK, Singh G, Singh SK, Karunanand B, Gunjan G, Agrawal SK. Exploring the link between leptin levels and metabolic syndrome in elderly Indian patients: Implications for family medicine and primary care practices. J Family Med Prim Care 2024; 13:3633-3638. [PMID: 39464951 PMCID: PMC11504790 DOI: 10.4103/jfmpc.jfmpc_2008_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/17/2024] [Accepted: 02/19/2024] [Indexed: 10/29/2024] Open
Abstract
Background The metabolic syndrome (MetS), according to the Adult Treatment Panel III of the National Cholesterol Education Programme, is a collection of metabolic abnormalities that includes one, two, or all three of the following traits: obesity in the abdomen, dyslipidemia, hypertension, fasting blood sugar, or insulin resistance. This study's aim was to assess the relationship between fasting serum leptin and MetS in elderly adults with T2DM in the Northern Indian population. Material and Methods The following information was collected from all the participants: (1) anthropometric data, (2) biochemical data, and (3) a lifestyle questionnaire on sociodemographic data, dietary practices, smoking, and alcohol intake to identify their risk factors for diabetes mellitus, CVD, and hypertension. Results A total of 36 older participants (56.30%) had a history of hypertension, while 29 elderly participants (44.61%) had diabetes mellitus. A total of 32 elderly participants (49.2%) had MetS, and this group had higher serum leptin (P 0.003), body weight (P = 0.019), BMI (P 0.001), waist circumference (P 0.001), CRP (P = 0.021), insulin (P = 0.001), and HOMA-IR (P = 0.003) values as well as higher percentages of females (P = 0.001), and those with type 2 diabetes mellitus (P = 0.002) and hypertension (P = 0.039) than those in the non-MetS group. Conclusion In older persons with T2DM, our study discovered a favorable correlation between serum leptin and MetS. It can act as a standalone indicator of MetS, offering a way to spot populations at risk for associated consequences and enabling early intervention.
Collapse
Affiliation(s)
- Arjun Kumar Singhal
- Department of Biochemistry, Faculty of Medicine and Health Sciences, SGT, Gurugram, Haryana, India
| | - Gaurav Singh
- Department of Microbiology, ESIC Medical College and Hospital, Patna, Bihar, India
| | - Shravan Kumar Singh
- Department of Medical Health and Family Welfare, Plibhit, Uttar Pradesh, India
| | - Busi Karunanand
- Department of Biochemistry, Faculty of Medicine and Health Sciences, SGT, Gurugram, Haryana, India
| | - Gagan Gunjan
- Department of General Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Sonu K. Agrawal
- Consultant Microbiologist, Vimta Labs. Limited, New Delhi, India
| |
Collapse
|
41
|
Li Z, Liang S, Ke L, Wang M, Gao K, Li D, Xu Z, Li N, Zhang P, Cheng W. Cell life-or-death events in osteoporosis: All roads lead to mitochondrial dynamics. Pharmacol Res 2024; 208:107383. [PMID: 39214266 DOI: 10.1016/j.phrs.2024.107383] [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] [Received: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria exhibit heterogeneous shapes and networks within and among cell types and tissues, also in normal or osteoporotic bone tissues with complex cell types. This dynamic characteristic is determined by the high plasticity provided by mitochondrial dynamics and is stemmed from responding to the survival and functional requirements of various bone cells in a specific microenvironments. In contrast, mitochondrial dysfunction, induced by dysregulation of mitochondrial dynamics, may act as a trigger of cell death signals, including common apoptosis and other forms of programmed cell death (PCD). These PCD processes consisting of tightly structured cascade gene expression events, can further influence the bone remodeling by facilitating the death of various bone cells. Mitochondrial dynamics, therefore, drive the bone cells to stand at the crossroads of life and death by integrating external signals and altering metabolism, shape, and signal-response properties of mitochondria. This implies that targeting mitochondrial dynamics displays significant potential in treatment of osteoporosis. Considerable effort has been made in osteoporosis to emphasize the parallel roles of mitochondria in regulating energy metabolism, calcium signal transduction, oxidative stress, inflammation, and cell death. However, the emerging field of mitochondrial dynamics-related PCD is not well understood. Herein, to bridge the gap, we outline the latest knowledge on mitochondrial dynamics regulating bone cell life or death during normal bone remodeling and osteoporosis.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050011, China
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000, China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300, China.
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
42
|
L'homme L, Sermikli BP, Haas JT, Fleury S, Quemener S, Guinot V, Barreby E, Esser N, Caiazzo R, Verkindt H, Legendre B, Raverdy V, Cheval L, Paquot N, Piette J, Legrand-Poels S, Aouadi M, Pattou F, Staels B, Dombrowicz D. Adipose tissue macrophage infiltration and hepatocyte stress increase GDF-15 throughout development of obesity to MASH. Nat Commun 2024; 15:7173. [PMID: 39169003 PMCID: PMC11339436 DOI: 10.1038/s41467-024-51078-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
Plasma growth differentiation factor-15 (GDF-15) levels increase with obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) but the underlying mechanism remains poorly defined. Using male mouse models of obesity and MASLD, and biopsies from carefully-characterized patients regarding obesity, type 2 diabetes (T2D) and MASLD status, we identify adipose tissue (AT) as the key source of GDF-15 at onset of obesity and T2D, followed by liver during the progression towards metabolic dysfunction-associated steatohepatitis (MASH). Obesity and T2D increase GDF15 expression in AT through the accumulation of macrophages, which are the main immune cells expressing GDF15. Inactivation of Gdf15 in macrophages reduces plasma GDF-15 concentrations and exacerbates obesity in mice. During MASH development, Gdf15 expression additionally increases in hepatocytes through stress-induced TFEB and DDIT3 signaling. Together, these results demonstrate a dual contribution of AT and liver to GDF-15 production in metabolic diseases and identify potential therapeutic targets to raise endogenous GDF-15 levels.
Collapse
Affiliation(s)
- Laurent L'homme
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| | - Benan Pelin Sermikli
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Joel T Haas
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Sébastien Fleury
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Sandrine Quemener
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Valentine Guinot
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Emelie Barreby
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Nathalie Esser
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU Liège, Liège, Belgium
| | - Robert Caiazzo
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Hélène Verkindt
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Benjamin Legendre
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Violeta Raverdy
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Lydie Cheval
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
- CNRS EMR 8228-Unité Métabolisme et Physiologie Rénale, Paris, France
| | - Nicolas Paquot
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU Liège, Liège, Belgium
| | - Jacques Piette
- Laboratory of Virology and Immunology, GIGA-Signal Transduction, University of Liège, Liège, Belgium
| | - Sylvie Legrand-Poels
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
| | - Myriam Aouadi
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - François Pattou
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Bart Staels
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - David Dombrowicz
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| |
Collapse
|
43
|
Luca T, Pezzino S, Puleo S, Castorina S. Lesson on obesity and anatomy of adipose tissue: new models of study in the era of clinical and translational research. J Transl Med 2024; 22:764. [PMID: 39143643 PMCID: PMC11323604 DOI: 10.1186/s12967-024-05547-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 07/28/2024] [Indexed: 08/16/2024] Open
Abstract
Obesity is a serious global illness that is frequently associated with metabolic syndrome. Adipocytes are the typical cells of adipose organ, which is composed of at least two different tissues, white and brown adipose tissue. They functionally cooperate, interconverting each other under physiological conditions, but differ in their anatomy, physiology, and endocrine functions. Different cellular models have been proposed to study adipose tissue in vitro. They are also useful for elucidating the mechanisms that are responsible for a pathological condition, such as obesity, and for testing therapeutic strategies. Each cell model has its own characteristics, culture conditions, advantages and disadvantages. The choice of one model rather than another depends on the specific study the researcher is conducting. In recent decades, three-dimensional cultures, such as adipose spheroids, have become very attractive because they more closely resemble the phenotype of freshly isolated cells. The use of such models has developed in parallel with the evolution of translational research, an interdisciplinary branch of the biomedical field, which aims to learn a scientific translational approach to improve human health and longevity. The focus of the present review is on the growing body of data linking the use of new cell models and the spread of translational research. Also, we discuss the possibility, for the future, to employ new three-dimensional adipose tissue cell models to promote the transition from benchside to bedsite and vice versa, allowing translational research to become routine, with the final goal of obtaining clinical benefits in the prevention and treatment of obesity and related disorders.
Collapse
Affiliation(s)
- Tonia Luca
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy.
| | | | - Stefano Puleo
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| |
Collapse
|
44
|
Lempicki MD, Gray JA, Abuna G, Murata RM, Divanovic S, McNamara CA, Meher AK. BAFF neutralization impairs the autoantibody-mediated clearance of dead adipocytes and aggravates obesity-induced insulin resistance. Front Immunol 2024; 15:1436900. [PMID: 39185417 PMCID: PMC11341376 DOI: 10.3389/fimmu.2024.1436900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
B cell-activating factor (BAFF) is a critical TNF-family cytokine that regulates homeostasis and peripheral tolerance of B2 cells. BAFF overproduction promotes autoantibody generation and autoimmune diseases. During obesity, BAFF is predominantly produced by white adipose tissue (WAT), and IgG autoantibodies against adipocytes are identified in the WAT of obese humans. However, it remains to be determined if the autoantibodies formed during obesity affect WAT remodeling and systemic insulin resistance. Here, we show that IgG autoantibodies are generated in high-fat diet (HFD)-induced obese mice that bind to apoptotic adipocytes and promote their phagocytosis by macrophages. Next, using murine models of obesity in which the gonadal WAT undergoes remodeling, we found that BAFF neutralization depleted IgG autoantibodies, increased the number of dead adipocytes, and exacerbated WAT inflammation and insulin resistance. RNA sequencing of the stromal vascular fraction from the WAT revealed decreased expression of immunoglobulin light-chain and heavy-chain variable genes suggesting a decreased repertoire of B cells after BAFF neutralization. Further, the B cell activation and the phagocytosis pathways were impaired in the WAT of BAFF-neutralized mice. In vitro, plasma IgG fractions from BAFF-neutralized mice reduced the phagocytic clearance of apoptotic adipocytes. Altogether, our study suggests that IgG autoantibodies developed during obesity, at least in part, dampens exacerbated WAT inflammation and systemic insulin resistance.
Collapse
Affiliation(s)
- Melissa D. Lempicki
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Jake A. Gray
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Gabriel Abuna
- School of Dental Medicine, East Carolina University, Greenville, NC, United States
| | - Ramiro M. Murata
- School of Dental Medicine, East Carolina University, Greenville, NC, United States
| | - Senad Divanovic
- Department of Pediatrics University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Coleen A. McNamara
- Cardiovascular Research Center, Cardiovascular Division, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Akshaya K. Meher
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| |
Collapse
|
45
|
Lanka N, Acharya P, Virani S, Afreen S, Perthiani A, Sangster E, Arcia Franchini AP. Safety and Efficacy of Canakinumab for the Prevention and Control of Type 2 Diabetes Mellitus and Its Complications: A Systematic Review. Cureus 2024; 16:e67065. [PMID: 39286685 PMCID: PMC11403928 DOI: 10.7759/cureus.67065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Today, diabetes mellitus (DM) is one of the leading causes of morbidity and mortality globally.In this grim context, while our current armamentarium of anti-diabetic agents is vast and increasingly available, glycemic control in a significant proportion of these patients continues to remain sub-optimal.This necessitates the exploration of other potential cellular pathways and targets to effectively manage this notorious disease and its numerous complications. Inflammatory responses are thought to be implicated in the decline of pancreatic beta-cell function, with interleukin-1 beta (IL-1β) playing an important role in these pathways. Canakinumab, a human monoclonal anti-IL-1β antibody, operates by reducing inflammation, potentially safeguarding or enhancing pancreatic beta-cell function. This systematic review aims to study the safety and efficacy of canakinumab in the prevention and control of type 2 diabetes mellitus (T2DM) and its complications. This study was conducted in accordance with the PRISMA 2020 Guidelines. PubMed including MEDLINE, Google Scholar and Cochrane Library were used as information sources and randomized clinical trials and retrospective observational studies evaluating patients with T2DM or impaired glucose tolerance with/without complications receiving canakinumab, compared with placebo or standard therapy and reporting about glycemic indicators including hemoglobin A1C (HbA1C) or blood sugar levels (BSL) or insulin levels and/or inflammatory indicators including high-sensitivity C-reactive protein (hsCRP) or interleukin-6 (IL-6) were included. Non-randomized clinical trials, animal studies, review articles, case reports, case series, studies not in the English language and those evaluating type 1 DM were excluded. In total, 271 studies were identified to be included in this study. Subsequently, 27 were found to be duplicate records and were eliminated. Manual screening of title/abstract of 244 records was done which found 207 to be ineligible and 37 studies were shortlisted. These were retrieved and full-text screening was undertaken which resulted in the exclusion of 28 reports due to the following reasons: ineligible study design (17), studies evaluating type 1 DM (three), studies evaluating anakinra (one), trial being canceled (three) and duplicate studies (four). Subsequently, a total of nine studies were included in the final review. All studies were included post quality appraisal. We found that canakinumab had a modest but mostly non-significant effect on glycemic parameters including HbA1C, while having a consistently significant reduction in systemic inflammatory parameters like hsCRP and IL-6. Additionally, it was found to have a significant reduction in incident major adverse cardiovascular events (MACE). Canakinumab was also found to be safe and well-tolerated in all patient populations. Although canakinumab did not reduce incident T2DM, an exploration of alternative pathways and targets implicated in the pathogenesis of this disease process is warranted for the prevention and control of T2DM.
Collapse
Affiliation(s)
- Nidhi Lanka
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Prakash Acharya
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Shikha Virani
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sumayya Afreen
- Obstetrics and Gynaecology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Arvin Perthiani
- General Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Elizabeth Sangster
- Psychiatry and Behavioral Sciences, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ana P Arcia Franchini
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
46
|
DeBari MK, Johnston EK, Scott JV, Ilzuka E, Sun W, Webster-Wood VA, Abbott RD. A Preliminary Study on Factors That Drive Patient Variability in Human Subcutaneous Adipose Tissues. Cells 2024; 13:1240. [PMID: 39120271 PMCID: PMC11311805 DOI: 10.3390/cells13151240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Adipose tissue is a dynamic regulatory organ that has profound effects on the overall health of patients. Unfortunately, inconsistencies in human adipose tissues are extensive and multifactorial, including large variability in cellular sizes, lipid content, inflammation, extracellular matrix components, mechanics, and cytokines secreted. Given the high human variability, and since much of what is known about adipose tissue is from animal models, we sought to establish correlations and patterns between biological, mechanical, and epidemiological properties of human adipose tissues. To do this, twenty-six independent variables were cataloged for twenty patients, which included patient demographics and factors that drive health, obesity, and fibrosis. A factorial analysis for mixed data (FAMD) was used to analyze patterns in the dataset (with BMI > 25), and a correlation matrix was used to identify interactions between quantitative variables. Vascular endothelial growth factor A (VEGFA) and actin alpha 2, smooth muscle (ACTA2) gene expression were the highest loadings in the first two dimensions of the FAMD. The number of adipocytes was also a key driver of patient-related differences, where a decrease in the density of adipocytes was associated with aging. Aging was also correlated with a decrease in overall lipid percentage of subcutaneous tissue, with lipid deposition being favored extracellularly, an increase in transforming growth factor-β1 (TGFβ1), and an increase in M1 macrophage polarization. An important finding was that self-identified race contributed to variance between patients in this study, where Black patients had significantly lower gene expression levels of TGFβ1 and ACTA2. This finding supports the urgent need to account for patient ancestry in biomedical research to develop better therapeutic strategies for all patients. Another important finding was that TGFβ induced factor homeobox 1 (TGIF1), an understudied signaling molecule, which is highly correlated with leptin signaling, was correlated with metabolic inflammation. Furthermore, this study draws attention to what we define as "extracellular lipid droplets", which were consistently found in collagen-rich regions of the obese adipose tissues evaluated here. Reduced levels of TGIF1 were correlated with higher numbers of extracellular lipid droplets and an inability to suppress fibrotic changes in adipose tissue. Finally, this study indicated that M1 and M2 macrophage markers were correlated with each other and leptin in patients with a BMI > 25. This finding supports growing evidence that macrophage polarization in obesity involves a complex, interconnecting network system rather than a full switch in activation patterns from M2 to M1 with increasing body mass. Overall, this study reinforces key findings in animal studies and identifies important areas for future research, where human and animal studies are divergent. Understanding key drivers of human patient variability is required to unravel the complex metabolic health of unique patients.
Collapse
Affiliation(s)
- Megan K. DeBari
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Elizabeth K. Johnston
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Jacqueline V. Scott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Erica Ilzuka
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Wenhuan Sun
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Victoria A. Webster-Wood
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Rosalyn D. Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| |
Collapse
|
47
|
Liang Y, Kaushal D, Wilson RB. Cellular Senescence and Extracellular Vesicles in the Pathogenesis and Treatment of Obesity-A Narrative Review. Int J Mol Sci 2024; 25:7943. [PMID: 39063184 PMCID: PMC11276987 DOI: 10.3390/ijms25147943] [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: 05/30/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
This narrative review explores the pathophysiology of obesity, cellular senescence, and exosome release. When exposed to excessive nutrients, adipocytes develop mitochondrial dysfunction and generate reactive oxygen species with DNA damage. This triggers adipocyte hypertrophy and hypoxia, inhibition of adiponectin secretion and adipogenesis, increased endoplasmic reticulum stress and maladaptive unfolded protein response, metaflammation, and polarization of macrophages. Such feed-forward cycles are not resolved by antioxidant systems, heat shock response pathways, or DNA repair mechanisms, resulting in transmissible cellular senescence via autocrine, paracrine, and endocrine signaling. Senescence can thus affect preadipocytes, mature adipocytes, tissue macrophages and lymphocytes, hepatocytes, vascular endothelium, pancreatic β cells, myocytes, hypothalamic nuclei, and renal podocytes. The senescence-associated secretory phenotype is closely related to visceral adipose tissue expansion and metaflammation; inhibition of SIRT-1, adiponectin, and autophagy; and increased release of exosomes, exosomal micro-RNAs, pro-inflammatory adipokines, and saturated free fatty acids. The resulting hypernefemia, insulin resistance, and diminished fatty acid β-oxidation lead to lipotoxicity and progressive obesity, metabolic syndrome, and physical and cognitive functional decline. Weight cycling is related to continuing immunosenescence and exposure to palmitate. Cellular senescence, exosome release, and the transmissible senescence-associated secretory phenotype contribute to obesity and metabolic syndrome. Targeted therapies have interrelated and synergistic effects on cellular senescence, obesity, and premature aging.
Collapse
Affiliation(s)
- Yicong Liang
- Bankstown Hospital, University of New South Wales, Sydney, NSW 2560, Australia;
| | - Devesh Kaushal
- Campbelltown Hospital, Western Sydney University, Sydney, NSW 2560, Australia;
| | - Robert Beaumont Wilson
- School of Clinical Medicine, University of New South Wales, High St., Kensington, Sydney, NSW 2052, Australia
| |
Collapse
|
48
|
Gheorghe AM, Ciobica ML, Nistor C, Gurzun MM, Sandulescu BA, Stanciu M, Popa FL, Carsote M. Inquiry of the Metabolic Traits in Relationship with Daily Magnesium Intake: Focus on Type 2 Diabetic Population. Clin Pract 2024; 14:1319-1347. [PMID: 39051301 PMCID: PMC11270223 DOI: 10.3390/clinpract14040107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024] Open
Abstract
Magnesium (Mg), an essential nutrient with a wide area of physiological roles, stands as a cofactor in over 600 enzymatic reactions involved in the synthesis of proteins and nucleic acids, DNA repair, neuromuscular functions, neuronal transmission, cardiac rhythm regulation, and the modulation of metabolic pathways, as well as acting as a natural blocker for the calcium channels. Our objective was to highlight the most recent clinical data with respect to daily Mg intake (DMI) and metabolic traits, particularly type 2 diabetes mellitus (DM). This was a PubMed-based review of the English-language medical papers across different key terms of search; the time frame was from January 2019 until April 2024. We included (clinically relevant) original studies and excluded cases reports, series, reviews, editorials, opinion, experimental studies, and non-human data as well as studies that did not specifically assessed DMI and only provided assays of serum Mg, studies on patients diagnosed with type 1 or secondary DM. A total of 30 studies were included and we organized the key findings into several sections as follows. Studies investigating DMI in relationship with the adherence to local recommendations in diabetic subjects (n = 2, one transversal and another retrospective cohort; N = 2823) found that most of them had lower DMI. Deficient DMI was correlated with the risk of developing/having DM across five studies (n = 5, one prospective and four of cross-sectional design; N = 47,166). An inverse correlation between DMI and DM prevalence was identified, but these data are presented amid a rather heterogeneous spectrum. Four novel studies (N = 7279) analysed the relationship between DMI and DM control according to various methods (HbA1c, fasting and postprandial glycaemia, and insulin); the association may be linear in diabetic subjects only at certain levels of DMI; additionally, the multifactorial influence on HBA1c should take into consideration this dietary determinant, as well, but there are no homogenous results. Three studies concerning DMI and diabetic complications (one cross-sectional, one prospective, and another case-control study) in terms of retinopathy (n = 1, N = 3794) and nephropathy (n = 2, N = 4805) suggested a lower DMI was associated with a higher risk of such complications. Additionally, two other studies (one prospective and one retrospective cohort) focused on mortality (N = 6744), which, taking only certain mortality indicators into consideration, might be decreased in the subgroups with a higher DMI. Seven studies (N = 30,610) analysed the perspective of DMI in the general population with the endpoint of different features amid glucose profile, particularly, insulin resistance. Concerning HOMA-IR, there were three confirmatory studies and one non-confirmatory, while fasting plasma glucose was highlighted as inversely correlated with a DMI (n = 1). The highest level of evidence regarding Mg supplementation effects on glucose metabolism stands on seven randomised controlled trials (N = 350). However, the sample size was reduced (from 14 to 86 individuals per study, either diabetic or pre-diabetic) and outcomes were rather discordant. These clinical aspects are essential from a multidisciplinary perspective and further trials are mandatory to address the current areas of discordant results.
Collapse
Affiliation(s)
- Ana-Maria Gheorghe
- PhD Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.G.); (B.-A.S.)
- Department of Clinical Endocrinology V, “C.I. Parhon” National Institute of Endocrinology, 011863 Bucharest, Romania;
| | - Mihai-Lucian Ciobica
- Department of Internal Medicine and Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Internal Medicine I and Rheumatology, “Dr. Carol Davila” Central Military University Emergency Hospital, 010825 Bucharest, Romania
| | - Claudiu Nistor
- Department 4-Cardio-Thoracic Pathology, Thoracic Surgery II Discipline, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Thoracic Surgery Department, “Dr. Carol Davila” Central Military University Emergency Hospital, 010242 Bucharest, Romania
| | - Maria-Magdalena Gurzun
- Cardiology Discipline, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Laboratory of Non-Invasive Cardiovascular Exploration, “Dr. Carol Davila” Central Military University Emergency Hospital, 010242 Bucharest, Romania
| | - Bianca-Andreea Sandulescu
- PhD Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.G.); (B.-A.S.)
- Department of Internal Medicine and Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Internal Medicine I and Rheumatology, “Dr. Carol Davila” Central Military University Emergency Hospital, 010825 Bucharest, Romania
| | - Mihaela Stanciu
- Department of Endocrinology, Faculty of Medicine, Lucian Blaga University of Sibiu, 550024 Sibiu, Romania;
| | - Florina Ligia Popa
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550024 Sibiu, Romania;
| | - Mara Carsote
- Department of Clinical Endocrinology V, “C.I. Parhon” National Institute of Endocrinology, 011863 Bucharest, Romania;
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
49
|
Savulescu-Fiedler I, Mihalcea R, Dragosloveanu S, Scheau C, Baz RO, Caruntu A, Scheau AE, Caruntu C, Benea SN. The Interplay between Obesity and Inflammation. Life (Basel) 2024; 14:856. [PMID: 39063610 PMCID: PMC11277997 DOI: 10.3390/life14070856] [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: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is an important condition affecting the quality of life of numerous patients and increasing their associated risk for multiple diseases, including tumors and immune-mediated disorders. Inflammation appears to play a major role in the development of obesity and represents a central point for the activity of cellular and humoral components in the adipose tissue. Macrophages play a key role as the main cellular component of the adipose tissue regulating the chronic inflammation and modulating the secretion and differentiation of various pro- and anti-inflammatory cytokines. Inflammation also involves a series of signaling pathways that might represent the focus for new therapies and interventions. Weight loss is essential in decreasing cardiometabolic risks and the degree of associated inflammation; however, the latter can persist for long after the excess weight is lost, and can involve changes in macrophage phenotypes that can ensure the metabolic adjustment. A clear understanding of the pathophysiological processes in the adipose tissue and the interplay between obesity and chronic inflammation can lead to a better understanding of the development of comorbidities and may ensure future targets for the treatment of obesity.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Razvan Mihalcea
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Radu Octavian Baz
- Clinical Laboratory of Radiology and Medical Imaging, “Sf. Apostol Andrei” County Emergency Hospital, 900591 Constanta, Romania
- Department of Radiology and Medical Imaging, Faculty of Medicine, “Ovidius” University, 900527 Constanta, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- “Prof. Dr. Matei Balș” National Institute for Infectious Diseases, 021105 Bucharest, Romania
| |
Collapse
|
50
|
Gogg S, Nerstedt A, Smith U, Hansson E. Breast volume in non-obese females is related to breast adipose cell hypertrophy, inflammation, and COX2 expression. J Plast Surg Hand Surg 2024; 59:83-88. [PMID: 38967364 DOI: 10.2340/jphs.v59.40754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/13/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Breast hypertrophy seems to be a risk factor for breast cancer and the amount and characteristics of breast adipose tissue may play important roles. The main aim of this study was to investigate associations between breast volume in normal weight women and hypertrophic adipose tissue and inflammation. METHODS Fifteen non-obese women undergoing breast reduction surgery were examined. Breast volume was measured with plastic cups and surgery was indicated if the breast was 800 ml or larger according to Swedish guidelines. We isolated adipose cells from the breasts and ambient subcutaneous tissue to measure cell size, cell inflammation and other known markers of risk of developing breast cancer including COX2 gene activation and MAPK, a cell proliferation regulator. RESULTS Breast adipose cell size was characterized by cell hypertrophy and closely related to breast volume. The breast adipose cells were also characterized by being pro-inflammatory with increased IL-6, IL-8, IL-1β, CCL-2, TNF-a and an increased marker of cell senescence GLB1/β-galactosidase, commonly increased in hypertrophic adipose tissue. The prostaglandin synthetic marker COX2 was also increased in the hypertrophic cells and COX2 has previously been shown to be an important marker of risk of developing breast cancer. Interestingly, the phosphorylation of the proliferation marker MAPK was also increased in the hypertrophic adipose cells. CONCLUSION Taken together, these findings show that increased breast volume in non-obese women is associated with adipose cell hypertrophy and dysfunction and characterized by increased inflammation and other markers of increased risk for developing breast cancer. TRIAL REGISTRATION Projektdatabasen FoU i VGR, project number: 249191 (https://www.researchweb.org/is/vgr/project/249191).
Collapse
Affiliation(s)
- Silvia Gogg
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Annika Nerstedt
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Emma Hansson
- Department of Plastic Surgery, Institute of Clinical Sciences, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Plastic Surgery, Gothenburg, Sweden.
| |
Collapse
|