1
|
Xu W, Zhang D, Ma Y, Gaspar RC, Kahn M, Nasiri A, Murray S, Samuel VT, Shulman GI. Ceramide synthesis inhibitors prevent lipid-induced insulin resistance through the DAG-PKCε-insulin receptor T1150 phosphorylation pathway. Cell Rep 2024; 43:114746. [PMID: 39302831 DOI: 10.1016/j.celrep.2024.114746] [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: 04/09/2024] [Revised: 07/22/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Abstract
Inhibition of the ceramide synthetic pathway with myriocin or an antisense oligonucleotide (ASO) targeting dihydroceramide desaturase (DES1) both improved hepatic insulin sensitivity in rats fed either a saturated or unsaturated fat diet and was associated with reductions in both hepatic ceramide and plasma membrane (PM)-sn-1,2-diacylglycerol (DAG) content. The insulin sensitizing effects of myriocin and Des1 ASO were abrogated by acute treatment with an ASO against DGAT2, which increased hepatic PM-sn-1,2-DAG but not hepatic C16 ceramide content. Increased PM-sn-1,2-DAG content was associated with protein kinase C (PKC)ε activation, increased insulin receptor (INSR)T1150 phosphorylation leading to reduced insulin-stimulated INSRY1152/AktS473 phosphorylation, and impaired insulin-mediated suppression of endogenous glucose production. These results demonstrate that inhibition of de novo ceramide synthesis by either myriocin treatment or DES1 knockdown protects against lipid-induced hepatic insulin resistance through a C16 ceramide-independent mechanism and that they mediate their effects to protect from lipid-induced hepatic insulin resistance via the PM-sn-1,2-DAG-PKCε-INSRT1150 phosphorylation pathway.
Collapse
Affiliation(s)
- Weiwei Xu
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Endocrinology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Dongyan Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yumin Ma
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Endocrinology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Rafael C Gaspar
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mario Kahn
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ali Nasiri
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sue Murray
- Ionis Pharmaceuticals, Carlsbad, CA 92010, USA
| | - Varman T Samuel
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; VA Connecticut Healthcare System, West Haven, CT 06516, USA.
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
2
|
Tang J, Lei Y, Pignalosa A, Hsu HH, Abdul-Sater AA, Sweeney G. Development of a non-invasive bioassay for adiponectin target engagement in mice. iScience 2024; 27:110994. [PMID: 39435143 PMCID: PMC11492082 DOI: 10.1016/j.isci.2024.110994] [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: 06/11/2024] [Revised: 07/23/2024] [Accepted: 09/16/2024] [Indexed: 10/23/2024] Open
Abstract
Adiponectin-based therapeutic strategies are promising for managing metabolic diseases and reducing inflammation, prompting the development of adiponectin receptor agonists. However, monitoring their pharmacodynamic actions in clinical applications is challenging. This study aimed to identify peripheral biomarkers to monitor adiponectin actions using ALY688, an adiponectin receptor agonist peptide. RNA sequencing analysis of whole blood identified a cluster of genes that were significantly increased in the ALY688-treated group compared to the control. This gene cluster was validated by qPCR and further confirmed in human peripheral blood mononuclear cells treated with ALY688 ex vivo. We also confirmed a functional outcome of ALY688 action in mice as our study also demonstrated the anti-inflammatory effect of ALY688 in a sublethal LPS mouse model. In summary, a newly identified gene cluster signature is suitable for assessing the pharmacodynamic action of adiponectin or its mimetics in blood samples.
Collapse
Affiliation(s)
- Jialing Tang
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Yubin Lei
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Angelica Pignalosa
- Allysta Pharmaceuticals Inc., 500 108th Avenue NE, Suite 1100, Bellevue, WA 98004, USA
| | - Henry H. Hsu
- Allysta Pharmaceuticals Inc., 500 108th Avenue NE, Suite 1100, Bellevue, WA 98004, USA
| | - Ali A. Abdul-Sater
- School of Kinesiology and Health Science, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Gary Sweeney
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
3
|
Xiao L, Mochizuki M, Shimamura N, Sunada K, Nakahara T. Interplay of co-cultured chimeric adipose and gingival tissues exacerbates inflammatory dysfunction relevant to periodontal and metabolic conditions. Life Sci 2024; 355:123009. [PMID: 39197574 DOI: 10.1016/j.lfs.2024.123009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024]
Abstract
Adipose tissue dysfunction is a key feature of metabolic syndrome, which increases the risk of periodontitis, an inflammatory disease induced by bacteria that affects the gingiva and other components of periodontal tissue. Recent studies indicate that molecules from inflamed periodontal tissue contribute to adipose tissue dysfunction. However, the cellular mechanisms and interactions between adipose tissue and gingiva driving the progression of metabolic and periodontal conditions remain unclear. To address this, we developed a chimeric (mouse/human) co-culture tissue model (which identifies the origins of species-specific cytokines) to investigate these interactions. Using tissue-specific functional cells and immunocytes, we constructed equivalents of adipose tissue (ATE) and gingiva (GTE), co-cultivating them under inflammatory conditions induced by bacterial endotoxin, lipopolysaccharide (LPS). Our findings showed that exposure to LPS resulted in a notable reduction in lipid accumulation, GLUT4 expression, and adiponectin secretion in ATE, along with increased macrophage colonies forming around lipid droplets, as well as elevated levels of triglyceride, leptin, and IL-6. In GTE, LPS triggered significant inflammatory responses, characterized by increased macrophage accumulation, elevated COX-2 expression, and heightened secretion of inflammatory cytokines. LPS also reduced epithelial thickness and the expression of keratin 19 and collagen IV, indicating impaired barrier function and gingival integrity. Co-culturing ATE with GTE exacerbated these LPS-induced harmful effects in both tissues. In conclusion, our findings suggest that interplay between gingiva and adipose tissue can intensify the inflammatory and dysfunctional changes caused by LPS. This co-culture tissue model offers a valuable tool for future studies on periodontitis and metabolic syndrome.
Collapse
Affiliation(s)
- Li Xiao
- Department of Physiology, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo 102-8159, Japan.
| | - Mai Mochizuki
- Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, Japan; Department of Life Science Dentistry, The Nippon Dental University, Japan.
| | - Naohiro Shimamura
- Department of Dental Anesthesiology, School of Life Dentistry at Tokyo, The Nippon Dental University, Japan.
| | - Katsuhisa Sunada
- Department of Dental Anesthesiology, School of Life Dentistry at Tokyo, The Nippon Dental University, Japan.
| | - Taka Nakahara
- Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, Japan.
| |
Collapse
|
4
|
LaMoia TE, Hubbard BT, Guerra MT, Nasiri A, Sakuma I, Kahn M, Zhang D, Goodman RP, Nathanson MH, Sancak Y, Perelis M, Mootha VK, Shulman GI. Cytosolic calcium regulates hepatic mitochondrial oxidation, intrahepatic lipolysis, and gluconeogenesis via CAMKII activation. Cell Metab 2024; 36:2329-2340.e4. [PMID: 39153480 PMCID: PMC11446666 DOI: 10.1016/j.cmet.2024.07.016] [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: 10/10/2023] [Revised: 05/06/2024] [Accepted: 07/19/2024] [Indexed: 08/19/2024]
Abstract
To examine the roles of mitochondrial calcium Ca2+ ([Ca2+]mt) and cytosolic Ca2+ ([Ca2+]cyt) in the regulation of hepatic mitochondrial fat oxidation, we studied a liver-specific mitochondrial calcium uniporter knockout (MCU KO) mouse model with reduced [Ca2+]mt and increased [Ca2+]cyt content. Despite decreased [Ca2+]mt, deletion of hepatic MCU increased rates of isocitrate dehydrogenase flux, α-ketoglutarate dehydrogenase flux, and succinate dehydrogenase flux in vivo. Rates of [14C16]palmitate oxidation and intrahepatic lipolysis were increased in MCU KO liver slices, which led to decreased hepatic triacylglycerol content. These effects were recapitulated with activation of CAMKII and abrogated with CAMKII knockdown, demonstrating that [Ca2+]cyt activation of CAMKII may be the primary mechanism by which MCU deletion promotes increased hepatic mitochondrial oxidation. Together, these data demonstrate that hepatic mitochondrial oxidation can be dissociated from [Ca2+]mt and reveal a key role for [Ca2+]cyt in the regulation of hepatic fat mitochondrial oxidation, intrahepatic lipolysis, gluconeogenesis, and lipid accumulation.
Collapse
Affiliation(s)
- Traci E LaMoia
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Brandon T Hubbard
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mateus T Guerra
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ali Nasiri
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ikki Sakuma
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mario Kahn
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Dongyan Zhang
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Russell P Goodman
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Michael H Nathanson
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yasemin Sancak
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | - Vamsi K Mootha
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Gerald I Shulman
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
5
|
Tan J, Li X, Dou N. Insulin Resistance Triggers Atherosclerosis: Caveolin 1 Cooperates with PKCzeta to Block Insulin Signaling in Vascular Endothelial Cells. Cardiovasc Drugs Ther 2024; 38:885-893. [PMID: 37289375 PMCID: PMC11438709 DOI: 10.1007/s10557-023-07477-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 06/09/2023]
Abstract
OBJECTIVE To date, therapies for endothelial dysfunction have primarily focused on ameliorating identified atherosclerosis (AS) risk factors rather than explicitly addressing endothelium-based mechanism. An in-depth exploration of the pathological mechanisms of endothelial injury was performed herein. METHODS Aortic caveolin 1 (Cav1) knockdown was achieved in mice using lentivirus, and AS was induced using a high-fat diet. Mouse body weight, blood glucose, insulin, lipid parameters, aortic plaque, endothelial injury, vascular nitric oxide synthase (eNOS), injury marker, and oxidative stress were examined. The effect of Cav1 knockdown on the content of PKCzeta and PI3K/Akt/eNOS pathway-related protein levels, as well as PKCzeta binding to Akt, was studied. ZIP, a PKCzeta inhibitor, was utilized to treat HUVECs in vitro, and the effect of ZIP on cell viability, inflammatory response, oxidative stress, and Akt activation was evaluated. RESULTS Cav1 knockdown had no significant effect on body weight or blood glucose in mice over an 8-week period, whereas drastically reduced insulin, lipid parameters, endothelial damage, E-selectin, and oxidative stress and elevated eNOS levels. Moreover, Cav1 knockdown triggered decreased PKCzeta enrichment and the activation of the PI3K/Akt/eNOS pathway. PKCzeta has a positive effect on cells without being coupled by Cav1, and ZIP had no marked influence on PKCzeta-Akt binding following Cav1/PKCzeta coupling. CONCLUSION Cav1/PKCzeta coupling antagonizes the activation of PI3K on Akt, leading to eNOS dysfunction, insulin resistance, and endothelial cell damage.
Collapse
Affiliation(s)
- Jingjing Tan
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200081, China
| | - Xiaoguang Li
- Department of Thyroid Breast and Vascular Surgery, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai, 200081, China
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai, 200003, China
| | - Ning Dou
- Department of Thyroid Breast and Vascular Surgery, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai, 200081, China.
| |
Collapse
|
6
|
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
|
7
|
Liu Y, Jiang H, Luo L, Gao Z. Relationship between four visceral obesity indices and prediabetes and diabetes: a cross-sectional study in Dalian, China. BMC Endocr Disord 2024; 24:191. [PMID: 39294627 PMCID: PMC11409536 DOI: 10.1186/s12902-024-01718-x] [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: 07/25/2024] [Accepted: 09/05/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND In recent times, a number of new indices for measuring visceral obesity have been developed. This research sought to investigate the relationship between four visceral obesity indices and prediabetes and diabetes. METHODS Conducted in 2011 as a cross-sectional analysis in Dalian, China, this study utilized logistic regression models to explore the relationships between four visceral obesity indices and prediabetes and diabetes. It also assessed the dose-response relationships using restricted cubic splines (RCS), performed subgroup analyses, and conducted interaction tests. The predictive values of four visceral obesity indices were evaluated using receiver operating characteristic (ROC) curves. RESULTS The study enrolled 10,090 participants, with prediabetes and diabetes prevalence at 80.53%. Multifactorial logistic regression revealed positive relationships between the four visceral obesity indices and prediabetes and diabetes. The results of the RCS analysis revealed a linear relationship between the body roundness index (BRI), cardiometabolic index (CMI) and prediabetes and diabetes. Conversely, a non-linear relationship was observed between Chinese visceral adiposity index (CVAI), lipid accumulation product (LAP) and prediabetes and diabetes. Subgroup analyses demonstrated stronger relationships of CMI, CVAI, and LAP with prediabetes and diabetes among females. ROC curves suggested that LAP could be an effective predictor of these conditions. CONCLUSION This research confirmed that four visceral obesity indices are linked with a higher risk of prediabetes and diabetes in middle-aged and elderly individuals in Dalian. Importantly, LAP could be an effective predictor of prediabetes and diabetes. Effective weight management significantly reduces the risk of both prediabetes and diabetes.
Collapse
Affiliation(s)
- Yuntong Liu
- Department of Endocrinology, Dalian Municipal Central Hospital, Dalian, China
- Dalian municipal Central Hospital, China Medical University, Shenyang, China
| | - Haodong Jiang
- Department of Cardiovascular, the Second Hospital of Jilin University, Changchun, China
| | - Lan Luo
- Department of Endocrinology, Dalian Municipal Central Hospital, Dalian, China.
| | - Zhengnan Gao
- Department of Endocrinology, Dalian Municipal Central Hospital, Dalian, China.
- Dalian municipal Central Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
8
|
Qu J, Fu S, Yin L, Zhang Q, Wang X. Chemerin influences blood lipid of aged male mice under high fat diet and exercise states through regulating the distribution and browning of white adipose tissue. Cytokine 2024; 181:156689. [PMID: 38981157 DOI: 10.1016/j.cyto.2024.156689] [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: 03/22/2024] [Revised: 05/13/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND With aging, white adipose tissue (WAT) undergoes distribution change and browning inhibition, which could be attenuated by exercise. Adipokine chemerin exerts roles in the above changes of WAT, and our previous studies demonstrated the effect of decreased chemerin on exercise-induced improvement of glucose and lipid metabolism in high fat diet (HFD) feeding male mice, so this study is to clarify whether chemerin's effects on glucose and lipid metabolism are associated with the distribution and browning of WAT. METHODS After diet and exercise interventions, body weight and adipose tissue contents in different depots of male mice were weighed, body composition and energy metabolism parameters were determined by Echo MRI Body Composition Analyzer and metabolic cage, respectively. The levels of serum adiponectin and leptin were detected by ELISA, and the protein levels of PGC-1α, UCP1, adiponectin and leptin in WAT were measured by Western blot. RESULTS Chemerin knockout exacerbated HFD-induced weight gain, upregulated the increases of visceral and subcutaneous WAT (vWAT and sWAT, especial in sWAT), and inhibited WAT browning, but improved blood lipid. Exercise reduced the body weight and WAT distribution, increased sWAT browning and further improved blood lipid in aged HFD male mice, which were abrogated by chemerin knockout. Detrimental alterations of leptin, adiponectin and adiponectin/leptin ratio were discovered in the serum and WAT of aged HFD chemerin(-/-) mice; and exercise-induced beneficial changes in these adipokines were blocked by chemerin knockout. CONCLUSION Chemerin influences blood lipid of aged male mice under HFD and exercise states through regulating the distribution and browning of WAT, which might be related to the changes of adiponectin, leptin and adiponectin/leptin ratio.
Collapse
Affiliation(s)
- Jing Qu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China; School of Physical Education, Minzu Normal University of Xingyi, Xingyi, Guizhou, China
| | - Shaoting Fu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China; Department of Kinesiology, College of Physical Education, Shanghai Normal University, Shanghai, China
| | - Lijun Yin
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Qilong Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Xiaohui Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
9
|
Sancar G, Birkenfeld AL. The role of adipose tissue dysfunction in hepatic insulin resistance and T2D. J Endocrinol 2024; 262:e240115. [PMID: 38967989 PMCID: PMC11378142 DOI: 10.1530/joe-24-0115] [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: 04/20/2024] [Accepted: 07/05/2024] [Indexed: 07/07/2024]
Abstract
The root cause of type 2 diabetes (T2D) is insulin resistance (IR), defined by the failure of cells to respond to circulating insulin to maintain lipid and glucose homeostasis. While the causes of whole-body insulin resistance are multifactorial, a major contributing factor is dysregulation of liver and adipose tissue function. Adipose dysfunction, particularly adipose tissue-IR (adipo-IR), plays a crucial role in the development of hepatic insulin resistance and the progression of metabolic dysfunction-associated steatotic liver disease (MASLD) in the context of T2D. In this review, we will focus on molecular mechanisms of hepatic insulin resistance and its association with adipose tissue function. A deeper understanding of the pathophysiological mechanisms of the transition from a healthy state to insulin resistance, impaired glucose tolerance, and T2D may enable us to prevent and intervene in the progression to T2D.
Collapse
Affiliation(s)
- Gencer Sancar
- German Center for Diabetes Research, Neuherberg, Germany
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- German Center for Diabetes Research, Neuherberg, Germany
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
10
|
Sannappa Gowda NG, Shiragannavar VD, Karunakara SH, Veeranna RP, Suvarna D, Kumar DP, Santhekadur PK. Novel role of Quercetin in ameliorating metabolic syndrome via VDR mediated activation of adiponectin/AdipoR2 signaling. Biochem Biophys Rep 2024; 39:101754. [PMID: 39006943 PMCID: PMC11246006 DOI: 10.1016/j.bbrep.2024.101754] [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: 04/11/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 07/16/2024] Open
Abstract
A sedentary lifestyle and physical inactivity leads to metabolic syndrome-associated comorbidities involving abdominal obesity, type 2 diabetes, hyperlipidaemia associated Cardiovascular Diseases (CVDs), and Metabolic dysfunction-associated fatty liver disease (MAFLD). In this study, we evaluated the novel hepato/cardio/adipo-protective role of Quercetin via Vitamin D Receptor, and elucidated its underlying mechanisms in reducing lipotoxicity, inflammation and fibrosis in high calorie diet induced metabolic syndrome. Male Swiss albino mice were fed with western diet and sugar water for multiple time intervals. Anti-lipotoxicity, anti-inflammatory, and anti-fibrotic effect of Quercetin was assessed by Oil Red O, H&E and TMS staining at different time points. The lipid profile, mRNA expression of inflammatory markers (TNF- α, IL-1β, IL-6 and MCP-1), fibrotic markers (α-SMA, COL1A1, COL1A2), adiponectin, AdipoR2, and VDR expression levels were measured from RNA pools of adipose, liver and heart tissues. Also, lipid-lowering and anti-steatohepatitic effects of Quercetin was assessed using mouse 3T3-L1 adipocytes, rat H9c2 cardiac cells, and human HepG2 hepatocytes. Our results indicate that, western diet fed mice with Quercetin ameliorated lipid profile and lipotoxicity. Histopathological examination and gene expression data revealed that Quercetin reduced hepatic and cardiac inflammation and fibrosis-associated markers. Interestingly, Quercetin treatment increased the serum levels of adiponectin and mRNA expressions of AdipoR2 and VDR. In-vitro experiments revealed the reduction in lipid accumulation of 3T3-L1 and fatty-acid-treated hepatic and cardiac cells following Quercetin treatment. These findings indicate that Quercetin exhibits a protective role on multiple organs through VDR activation and subsequent Adipo/AdipoR2 signaling in metabolic syndrome associated obesity, hepatic injury, and cardiac dysfunction.
Collapse
Affiliation(s)
- Nirmala G Sannappa Gowda
- Department of Biochemistry, Center of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Varsha D Shiragannavar
- Department of Biochemistry, Center of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Shreyas H Karunakara
- Department of Biochemistry, Center of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | | | - Deepak Suvarna
- Department of Gastroenterology, JSS Medical College and Hospital, JSS Academy of Higher Education and Research, Mysuru, 570004, India
| | - Divya P Kumar
- Department of Biochemistry, Center of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Prasanna K Santhekadur
- Department of Biochemistry, Center of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| |
Collapse
|
11
|
Toksoy Z, Ma Y, Goedeke L, Li W, Hu X, Wu X, Cacheux M, Liu Y, Akar FG, Shulman GI, Young LH. Role of AMPK in Atrial Metabolic Homeostasis and Substrate Preference. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.608789. [PMID: 39257756 PMCID: PMC11383699 DOI: 10.1101/2024.08.29.608789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Atrial fibrillation is the most common clinical arrhythmia and may be due in part to metabolic stress. Atrial specific deletion of the master metabolic sensor, AMP-activated protein kinase (AMPK), induces atrial remodeling culminating in atrial fibrillation in mice, implicating AMPK signaling in the maintenance of atrial electrical and structural homeostasis. However, atrial substrate preference for mitochondrial oxidation and the role of AMPK in regulating atrial metabolism are unknown. Here, using LC-MS/MS methodology combined with infusions of [ 13 C 6 ]glucose and [ 13 C 4 ]β-hydroxybutyrate in conscious mice, we demonstrate that conditional deletion of atrial AMPK catalytic subunits shifts mitochondrial atrial metabolism away from fatty acid oxidation and towards pyruvate oxidation. LC-MS/MS-based quantification of acyl-CoAs demonstrated decreased atrial tissue content of long-chain fatty acyl-CoAs. Proteomic analysis revealed a broad downregulation of proteins responsible for fatty acid uptake (LPL, CD36, FABP3), acylation and oxidation. Atrial AMPK deletion reduced expression of atrial PGC1-α and downstream PGC1-α/PPARα/RXR regulated gene transcripts. In contrast, atrial [ 14 C]2-deoxyglucose uptake and GLUT1 expression increased with fasting in mice with AMPK deletion, while the expression of glycolytic enzymes exhibited heterogenous changes. Thus, these results highlight the crucial homeostatic role of AMPK in the atrium, with loss of atrial AMPK leading to downregulation of the PGC1-α/PPARα pathway and broad metabolic reprogramming with a loss of fatty acid oxidation, which may contribute to atrial remodeling and arrhythmia.
Collapse
|
12
|
Liu QK. Mechanisms of action and therapeutic applications of GLP-1 and dual GIP/GLP-1 receptor agonists. Front Endocrinol (Lausanne) 2024; 15:1431292. [PMID: 39114288 PMCID: PMC11304055 DOI: 10.3389/fendo.2024.1431292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are two incretins that bind to their respective receptors and activate the downstream signaling in various tissues and organs. Both GIP and GLP-1 play roles in regulating food intake by stimulating neurons in the brain's satiety center. They also stimulate insulin secretion in pancreatic β-cells, but their effects on glucagon production in pancreatic α-cells differ, with GIP having a glucagonotropic effect during hypoglycemia and GLP-1 exhibiting glucagonostatic effect during hyperglycemia. Additionally, GIP directly stimulates lipogenesis, while GLP-1 indirectly promotes lipolysis, collectively maintaining healthy adipocytes, reducing ectopic fat distribution, and increasing the production and secretion of adiponectin from adipocytes. Together, these two incretins contribute to metabolic homeostasis, preventing both hyperglycemia and hypoglycemia, mitigating dyslipidemia, and reducing the risk of cardiovascular diseases in individuals with type 2 diabetes and obesity. Several GLP-1 and dual GIP/GLP-1 receptor agonists have been developed to harness these pharmacological effects in the treatment of type 2 diabetes, with some demonstrating robust effectiveness in weight management and prevention of cardiovascular diseases. Elucidating the underlying cellular and molecular mechanisms could potentially usher in the development of new generations of incretin mimetics with enhanced efficacy and fewer adverse effects. The treatment guidelines are evolving based on clinical trial outcomes, shaping the management of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Qiyuan Keith Liu
- MedStar Medical Group, MedStar Montgomery Medical Center, Olney, MD, United States
| |
Collapse
|
13
|
Fu L, Du J, Furkert D, Shipton ML, Liu X, Aguirre T, Chin AC, Riley AM, Potter BVL, Fiedler D, Zhang X, Zhu Y, Fu C. Depleting inositol pyrophosphate 5-InsP7 protected the heart against ischaemia-reperfusion injury by elevating plasma adiponectin. Cardiovasc Res 2024; 120:954-970. [PMID: 38252884 PMCID: PMC11218692 DOI: 10.1093/cvr/cvae017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 01/24/2024] Open
Abstract
AIMS Adiponectin is an adipocyte-derived circulating protein that exerts cardiovascular and metabolic protection. Due to the futile degradation of endogenous adiponectin and the challenges of exogenous administration, regulatory mechanisms of adiponectin biosynthesis are of significant pharmacological interest. METHODS AND RESULTS Here, we report that 5-diphosphoinositol 1,2,3,4,6-pentakisphosphate (5-InsP7) generated by inositol hexakisphosphate kinase 1 (IP6K1) governed circulating adiponectin levels via thiol-mediated protein quality control in the secretory pathway. IP6K1 bound to adiponectin and DsbA-L and generated 5-InsP7 to stabilize adiponectin/ERp44 and DsbA-L/Ero1-Lα interactions, driving adiponectin intracellular degradation. Depleting 5-InsP7 by either IP6K1 deletion or pharmacological inhibition blocked intracellular adiponectin degradation. Whole-body and adipocyte-specific deletion of IP6K1 boosted plasma adiponectin levels, especially its high molecular weight forms, and activated AMPK-mediated protection against myocardial ischaemia-reperfusion injury. Pharmacological inhibition of 5-InsP7 biosynthesis in wild-type but not adiponectin knockout mice attenuated myocardial ischaemia-reperfusion injury. CONCLUSION Our findings revealed that 5-InsP7 is a physiological regulator of adiponectin biosynthesis that is amenable to pharmacological intervention for cardioprotection.
Collapse
Affiliation(s)
- Lin Fu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Jimin Du
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - David Furkert
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Megan L Shipton
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Xiaoqi Liu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Tim Aguirre
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Alfred C Chin
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Andrew M Riley
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Barry V L Potter
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Dorothea Fiedler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Chenglai Fu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Yangpu District, Shanghai 200092, China
| |
Collapse
|
14
|
Mladenović D, Vesković M, Šutulović N, Hrnčić D, Stanojlović O, Radić L, Macut JB, Macut D. Adipose-derived extracellular vesicles - a novel cross-talk mechanism in insulin resistance, non-alcoholic fatty liver disease, and polycystic ovary syndrome. Endocrine 2024; 85:18-34. [PMID: 38285412 DOI: 10.1007/s12020-024-03702-w] [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: 11/17/2023] [Accepted: 01/14/2024] [Indexed: 01/30/2024]
Abstract
Obesity is the best described risk factor for the development of non-alcoholic fatty liver disease (NAFLD)/metabolic dysfunction associated steatotic liver disease (MASLD) and polycystic ovary syndrome (PCOS) while the major pathogenic mechanism linking these entities is insulin resistance (IR). IR is primarily caused by increased secretion of proinflammatory cytokines, adipokines, and lipids from visceral adipose tissue. Increased fatty acid mobilization results in ectopic fat deposition in the liver which causes endoplasmic reticulum stress, mitochondrial dysfunction, and oxidative stress resulting in increased cytokine production and subsequent inflammation. Similarly, IR with hyperinsulinemia cause hyperandrogenism, the hallmark of PCOS, and inflammation in the ovaries. Proinflammatory cytokines from both liver and ovaries aggravate IR thus providing a complex interaction between adipose tissue, liver, and ovaries in inducing metabolic abnormalities in obese subjects. Although many pathogenic mechanisms of IR, NAFLD/MASLD, and PCOS are known, there is still no effective therapy for these entities suggesting the need for further evaluation of their pathogenesis. Extracellular vesicles (EVs) represent a novel cross-talk mechanism between organs and include membrane-bound vesicles containing proteins, lipids, and nucleic acids that may change the phenotype and function of target cells. Adipose tissue releases EVs that promote IR, the development of all stages of NAFLD/MASLD and PCOS, while mesenchymal stem cell-derived AVs may alleviate metabolic abnormalities and may represent a novel therapeutic device in NAFLD/MASLD, and PCOS. The purpose of this review is to summarize the current knowledge on the role of adipose tissue-derived EVs in the pathogenesis of IR, NAFLD/MASLD, and PCOS.
Collapse
Affiliation(s)
- Dušan Mladenović
- Institute of Pathophysiology "Ljubodrag Buba Mihailovic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| | - Milena Vesković
- Institute of Pathophysiology "Ljubodrag Buba Mihailovic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nikola Šutulović
- Laboratory for Neurophysiology, Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragan Hrnčić
- Laboratory for Neurophysiology, Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Olivera Stanojlović
- Laboratory for Neurophysiology, Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Lena Radić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Jelica Bjekić Macut
- University of Belgrade Faculty of Medicine, Department of Endocrinology, UMC Bežanijska kosa, Belgrade, Serbia
| | - Djuro Macut
- University of Belgrade Faculty of Medicine, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Belgrade, Serbia
| |
Collapse
|
15
|
Frances L, Croyal M, Ruidavets JB, Maraninchi M, Combes G, Raffin J, de Souto Barreto P, Ferrières J, Blaak EE, Perret B, Moro C, Valéro R, Martinez LO, Viguerie N. Identification of circulating apolipoprotein M as a new determinant of insulin sensitivity and relationship with adiponectin. Int J Obes (Lond) 2024; 48:973-980. [PMID: 38491190 PMCID: PMC11216985 DOI: 10.1038/s41366-024-01510-w] [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: 11/06/2023] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND The adiponectin is one of the rare adipokines down-regulated with obesity and protects against obesity-related disorders. Similarly, the apolipoprotein M (apoM) is expressed in adipocytes and its expression in adipose tissue is associated with metabolic health. We compared circulating apoM with adiponectin regarding their relationship with metabolic parameters and insulin sensitivity and examined their gene expression patterns in adipocytes and in the adipose tissue. METHODS Circulating apoM and adiponectin were examined in 169 men with overweight in a cross-sectional study, and 13 patients with obesity during a surgery-induced slimming program. Correlations with clinical parameters including the insulin resistance index (HOMA-IR) were analyzed. Multiple regression analyses were performed on HOMA-IR. The APOM and ADIPOQ gene expression were measured in the adipose tissue from 267 individuals with obesity and a human adipocyte cell line. RESULTS Participants with type 2 diabetes had lower circulating adiponectin and apoM, while apoM was higher in individuals with dyslipidemia. Similar to adiponectin, apoM showed negative associations with HOMA-IR and hs-CRP (r < -0.2), and positive correlations with HDL markers (HDL-C and apoA-I, r > 0.3). Unlike adiponectin, apoM was positively associated with LDL markers (LDL-C and apoB100, r < 0.20) and negatively correlated with insulin and age (r < -0.2). The apoM was the sole negative determinant of HOMA-IR in multiple regression models, while adiponectin not contributing significantly. After surgery, the change in HOMA-IR was negatively associated with the change in circulating apoM (r = -0.71), but not with the change in adiponectin. The APOM and ADIPOQ gene expression positively correlated in adipose tissue (r > 0.44) as well as in adipocytes (r > 0.81). In adipocytes, APOM was downregulated by inflammatory factors and upregulated by adiponectin. CONCLUSIONS The apoM rises as a new partner of adiponectin regarding insulin sensitivity. At the adipose tissue level, the adiponectin may be supported by apoM to promote a healthy adipose tissue. TRIAL REGISTRATION NCT01277068, registered 13 January 2011; NCT02332434, registered 5 January 2015; and NCT00390637, registered 20 October 2006.
Collapse
Affiliation(s)
- Laurie Frances
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), UMR1297, 31432, Toulouse, France
| | - Mikaël Croyal
- Nantes Université, CHU Nantes, CNRS, INSERM, BioCore, US16, SFR Bonamy, 44000, Nantes, France
- CRNH-Ouest Mass Spectrometry Core Facility, 44000, Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'Institut du Thorax, 44000, Nantes, France
| | | | - Marie Maraninchi
- Aix Marseille Université, APHM, INSERM, INRAe, C2VN, Department of Nutrition, Metabolic Diseases and Endocrinology, University Hospital La Conception, 13385, Marseille, France
| | - Guillaume Combes
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), UMR1297, 31432, Toulouse, France
- Institut Hospitalo-Universitaire HealthAge, IHU HealthAge, Inserm, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
| | - Jérémy Raffin
- Institut Hospitalo-Universitaire HealthAge, IHU HealthAge, Inserm, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, 31000, Toulouse, France
| | - Philippe de Souto Barreto
- CERPOP UMR 1295, University of Toulouse III, Inserm, UPS, 31000, Toulouse, France
- Institut Hospitalo-Universitaire HealthAge, IHU HealthAge, Inserm, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, 31000, Toulouse, France
| | - Jean Ferrières
- CERPOP UMR 1295, University of Toulouse III, Inserm, UPS, 31000, Toulouse, France
- Department of Cardiology, Toulouse Rangueil University Hospital, Toulouse University School of Medicine, Toulouse, France
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+(MUMC+), Maastricht, The Netherlands
| | - Bertrand Perret
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), UMR1297, 31432, Toulouse, France
- Institut Hospitalo-Universitaire HealthAge, IHU HealthAge, Inserm, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
| | - Cédric Moro
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), UMR1297, 31432, Toulouse, France
| | - René Valéro
- Aix Marseille Université, APHM, INSERM, INRAe, C2VN, Department of Nutrition, Metabolic Diseases and Endocrinology, University Hospital La Conception, 13385, Marseille, France
| | - Laurent O Martinez
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), UMR1297, 31432, Toulouse, France.
- Institut Hospitalo-Universitaire HealthAge, IHU HealthAge, Inserm, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France.
| | - Nathalie Viguerie
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), UMR1297, 31432, Toulouse, France.
| |
Collapse
|
16
|
Fayazi F, Kheirouri S, Alizadeh M. Exploring effects of melatonin supplementation on insulin resistance: An updated systematic review of animal and human studies. Diabetes Metab Syndr 2024; 18:103073. [PMID: 39096757 DOI: 10.1016/j.dsx.2024.103073] [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: 04/07/2023] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 08/05/2024]
Abstract
BACKGROUND Insulin resistance (IR), defined as an impaired response to insulin stimulation of target tissues, is a substantial determinant of many metabolic disorders. This study aimed to update the findings of the previous systematic review evidence regarding the effect of melatonin on factors related to IR, including hyperinsulinemia, hyperglycemia, homeostasis model assessment of insulin resistance (HOMA-IR), and quantitative insulin sensitivity check index (QUICKI). METHODS We systematically reviewed the evidence on the impact of melatonin supplementation on IR indices, fasting insulin, and fasting plasma glucose. PubMed, ScienceDirect, SCOPUS, and Google Scholar databases were searched until March 2024. RESULTS We identified 6114 potentially relevant articles during the search. Eighteen animal studies and 15 randomized clinical trials met the inclusion criteria. The results indicated that melatonin supplementation reduced fasting plasma glucose (FPG, 14 out of 29 studies), fasting insulin (22 out of 28 studies), HOMA-IR (28 out of 33 studies), and increased QUICKI (7 out of 7 studies). According to RCT studies, melatonin treatment at a dosage of 10 mg reduced HOMA-IR levels in individuals with various health conditions. CONCLUSION According to most evidence, melatonin supplementation may decrease fasting insulin and HOMA-IR and increase QUICKI but may not affect FPG.
Collapse
Affiliation(s)
- Fakhrosadat Fayazi
- Student Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Sorayya Kheirouri
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Alizadeh
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
17
|
Cordero OJ, Kotrulev M, Gomez-Touriño I. Comment on Lai et al. Dipeptidyl Peptidase 4 Stimulation Induces Adipogenesis-Related Gene Expression of Adipose Stromal Cells. Int. J. Mol. Sci. 2023, 24, 16101. Int J Mol Sci 2024; 25:7093. [PMID: 39000199 PMCID: PMC11241282 DOI: 10.3390/ijms25137093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Adiponectin is a circulating hormone secreted by adipose tissue that exerts, unlike other adipokines such as leptin, anti-inflammatory, anti-atherosclerotic and other protective effects on health. Adiponectin receptor agonists are being tested in clinical trials and are expected to show benefits in many diseases. In a recent article, LW Chen's group used monocyte chemoattractant protein-1 (MCP-1/CCL2) to improve plasma levels of adiponectin, suggesting the involvement of dipeptidyl peptidase 4 (DPP4/CD26) in the mechanism. Here, we discuss the significance of the role of DPP4, favoring the increase in DPP4-positive interstitial progenitor cells, a finding that fits with the greater stemness and persistence of other DPP4/CD26-positive cells.
Collapse
Affiliation(s)
- Oscar J Cordero
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.K.); (I.G.-T.)
| | - Martin Kotrulev
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.K.); (I.G.-T.)
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Iria Gomez-Touriño
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.K.); (I.G.-T.)
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
18
|
Cai JJ, Zheng P, Su M, Shen YL, Li XC, Guo QW, Chen X, Su GM, Lin J, Gong RR, Fang DZ. Suicidal ideation in adolescents with adiponectin receptor 2 rs12342 polymorphism affected by Wenchuan earthquake. Early Interv Psychiatry 2024. [PMID: 38837550 DOI: 10.1111/eip.13585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
PURPOSE The present study was to investigate prevalence of suicidal ideation and its associations with biological and environmental factors in adolescents with different genotypes of rs12342 at adiponectin receptor 2 gene (ADIPOR2). METHODS Suicidal ideation, biological and environmental factors were evaluated by questionnaires in 669 high school students after Wenchuan earthquake in China. ADIPOR2 rs12342 was genotyped by polymerase chain reaction-restriction fragment length polymorphism and verified by DNA sequencing. RESULTS Female adolescents had higher prevalence of suicidal ideation than male students in AG heterozygote and GG homozygote, but not AA homozygote. Prevalence of suicidal ideation was different in male, but not female, subjects with different genotypes. Genotype and allele frequencies were significantly different between male students with and without suicidal ideation, but not the female counterparts. Family history of mental disorders, extent of damage to property, carbohydrate intake and protein intake were associated with suicidal ideation in female subjects, while ADIPOR2 rs12342, father's educational level and previous trauma experience were associated with suicidal ideation in male subjects. CONCLUSION ADIPOR2 rs12342 is associated with and has potential to interact with environmental factors on suicidal ideation in a gender-dependent manner in youth. These findings pave a novel way and perspective for precision inferences of suicidal ideation in subjects with different genetic backgrounds. ADIPOR2 rs12342 needs to be considered when intervening suicidal ideation, especially in adolescents.
Collapse
Affiliation(s)
- Jia Jing Cai
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Ping Zheng
- West China School of Nursing, Sichuan University, Chengdu, People's Republic of China
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Mi Su
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Yi Lin Shen
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Xue Cheng Li
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Qi Wei Guo
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Xu Chen
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Guo Ming Su
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Jia Lin
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Ren Rong Gong
- West China School of Nursing, Sichuan University, Chengdu, People's Republic of China
- Department of Surgery, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Ding Zhi Fang
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
19
|
Zhang M, Yue X, Xu S, Piao J, Zhao L, Shu S, Kuzuya M, Li P, Hong L, Kim W, Liu B, Cheng XW. Dipeptidyl peptidase-4 disturbs adipocyte differentiation via the negative regulation of the glucagon-like peptide-1/adiponectin-cathepsin K axis in mice under chronic stress conditions. FASEB J 2024; 38:e23684. [PMID: 38795334 DOI: 10.1096/fj.202400158r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/24/2024] [Accepted: 05/07/2024] [Indexed: 05/27/2024]
Abstract
Exposure to chronic psychosocial stress is a risk factor for metabolic disorders. Because dipeptidyl peptidase-4 (DPP4) and cysteinyl cathepsin K (CTSK) play important roles in human pathobiology, we investigated the role(s) of DPP4 in stress-related adipocyte differentiation, with a focus on the glucagon-like peptide-1 (GLP-1)/adiponectin-CTSK axis in vivo and in vitro. Plasma and inguinal adipose tissue from non-stress wild-type (DPP4+/+), DPP4-knockout (DPP4-/-) and CTSK-knockout (CTSK-/-) mice, and stressed DPP4+/+, DPP4-/-, CTSK-/-, and DPP4+/+ mice underwent stress exposure plus GLP-1 receptor agonist exenatide loading for 2 weeks and then were analyzed for stress-related biological and/or morphological alterations. On day 14 under chronic stress, stress decreased the weights of adipose tissue and resulted in harmful changes in the plasma levels of DPP4, GLP-1, CTSK, adiponectin, and tumor necrosis factor-α proteins and the adipose tissue levels of CTSK, preadipocyte factor-1, fatty acid binding protein-4, CCAAT/enhancer binding protein-α, GLP-1 receptor, peroxisome proliferator-activated receptor-γ, perilipin2, secreted frizzled-related protein-4, Wnt5α, Wnt11 and β-catenin proteins and/or mRNAs as well as macrophage infiltration in adipose tissue; these changes were rectified by DPP4 deletion. GLP-1 receptor activation and CTSK deletion mimic the adipose benefits of DPP4 deficiency. In vitro, CTSK silencing and overexpression respectively prevented and facilitated stress serum and oxidative stress-induced adipocyte differentiation accompanied with changes in the levels of pref-1, C/EBP-α, and PPAR-γ in 3T3-L1 cells. Thus, these findings indicated that increased DPP4 plays an essential role in stress-related adipocyte differentiation, possibly through a negative regulation of GLP-1/adiponectin-CTSK axis activation in mice under chronic stress conditions.
Collapse
Affiliation(s)
- Meiping Zhang
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, P. R. China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, P. R. China
| | - Xueling Yue
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, P. R. China
| | - Shengnan Xu
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, P. R. China
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Jinshun Piao
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, P. R. China
| | - Longguo Zhao
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, P. R. China
| | - Shangzhi Shu
- Department of Cardiovascular Disease, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Masafumi Kuzuya
- Department of Community Health & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ping Li
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Lan Hong
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, Jilin, P. R. China
| | - Weon Kim
- Department of Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, P. R. China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, P. R. China
| |
Collapse
|
20
|
El-Araby RE, Tu Q, Xie Y, Aboushousha T, Li Z, Xu X, Zhu ZX, Dong LQ, Chen J. Adiponectin mRNA Conjugated with Lipid Nanoparticles Specifically Targets the Pathogenesis of Type 2 Diabetes. Aging Dis 2024:AD.2024.0162. [PMID: 38916734 DOI: 10.14336/ad.2024.0162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/15/2024] [Indexed: 06/26/2024] Open
Abstract
Type 2 diabetes (T2D) is a widespread health condition both in the United States and around the world, with insulin resistance playing a critical role in its development. Effective treatment strategies are essential for managing T2D and mitigating associated risks. Adiponectin (APN), secreted by adipocytes, exhibits an inverse correlation with obesity-related adiposity, and its levels are negatively associated with insulin resistance and body mass index. This study aimed to enhance endogenous APN levels in a diet-induced obese (DIO) mouse model using lipid nanoparticles (LNP) as safe delivery agents for APN mRNA conjugates. The results indicate that APN-mRNA-LNP administration successfully induced APN synthesis in various tissues, including muscle, liver, kidney, pancreas, and adipose cells. This induction was associated with several positive outcomes, such as preventing diet-induced body weight gain, improving hyperglycemia by promoting Glut-4 expression, alleviating diabetic nephropathy symptoms by blocking the EGFR pathway, and reducing pro-inflammatory cytokine production. In addition, the treatment demonstrated enhanced insulin sensitivity by activating DGKd and inhibiting PKCε. This resulted in reactivation of insulin receptors in insulin target tissues and stimulation of insulin secretion from pancreatic beta cells. The findings of the present study highlight the potential of APN-mRNA-LNP-based nucleic acid therapy as a treatment for type 2 diabetes, offering a comprehensive approach to addressing its complexities.
Collapse
Affiliation(s)
- Rady E El-Araby
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- Theodor Bilharz Research Institute, Ministry of scientific Research, Cairo, Egypt
| | - Qisheng Tu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Ying Xie
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Tarek Aboushousha
- Theodor Bilharz Research Institute, Ministry of scientific Research, Cairo, Egypt
| | - Zhongyu Li
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Xiaoyang Xu
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Zoe X Zhu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Lily Q Dong
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, Texas 78229, USA
| | - Jake Chen
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- Department of Genetics, Molecular and Cellular Biology, Tufts University School of Medicine, and Graduate School of Biomedical Sciences. 136 Harrison Ave, M&;ampV 811, Boston, MA 02111, USA
| |
Collapse
|
21
|
Zhang S, Gao L, Li S, Luo M, Chen L, Xi Q, Zhao Z, Zhao Q, Yang T, Zeng Q, Li X, Huang Z, Duan A, Wang Y, Luo Q, Guo Y, Liu Z. Association of non-insulin-based insulin resistance indices with disease severity and adverse outcome in idiopathic pulmonary arterial hypertension: a multi-center cohort study. Cardiovasc Diabetol 2024; 23:154. [PMID: 38702735 PMCID: PMC11069206 DOI: 10.1186/s12933-024-02236-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Insulin resistance (IR) plays an important role in the pathophysiology of cardiovascular disease. Recent studies have shown that diabetes mellitus and impaired lipid metabolism are associated with the severity and prognosis of idiopathic pulmonary arterial hypertension (IPAH). However, the relationship between IR and pulmonary hypertension is poorly understood. This study explored the association between four IR indices and IPAH using data from a multicenter cohort. METHODS A total of 602 consecutive participants with IPAH were included in this study between January 2015 and December 2022. The metabolic score for IR (METS-IR), triglyceride to high-density lipoprotein cholesterol (TG/HDL-C) ratio, triglyceride and glucose (TyG) index, and triglyceride-glucose-body mass index (TyG-BMI) were used to quantify IR levels in patients with IPAH. The correlation between non-insulin-based IR indices and long-term adverse outcomes was determined using multivariate Cox regression models and restricted cubic splines. RESULTS During a mean of 3.6 years' follow-up, 214 participants experienced all-cause death or worsening condition. Compared with in low to intermediate-low risk patients, the TG/HDL-C ratio (2.9 ± 1.7 vs. 3.3 ± 2.1, P = 0.003) and METS-IR (34.5 ± 6.7 vs. 36.4 ± 7.5, P < 0.001) were significantly increased in high to intermediate-high risk patients. IR indices correlated with well-validated variables that reflected the severity of IPAH, such as the cardiac index and stroke volume index. Multivariate Cox regression analyses indicated that the TyG-BMI index (hazard ratio [HR] 1.179, 95% confidence interval [CI] 1.020, 1.363 per 1.0-standard deviation [SD] increment, P = 0.026) and METS-IR (HR 1.169, 95% CI 1.016, 1.345 per 1.0-SD increment, P = 0.030) independently predicted adverse outcomes. Addition of the TG/HDL-C ratio and METS-IR significantly improved the reclassification and discrimination ability beyond the European Society of Cardiology (ESC) risk score. CONCLUSIONS IR is associated with the severity and long-term prognosis of IPAH. TyG-BMI and METS-IR can independently predict clinical worsening events, while METS-IR also provide incremental predictive performance beyond the ESC risk stratification.
Collapse
Affiliation(s)
- Sicheng Zhang
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China
| | - Luyang Gao
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China
| | - Sicong Li
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China
| | - Manqing Luo
- Department of Cardiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, No. 134, East Street, Gulou District, Fuzhou, 350001, Fujian, China
| | - Lichuan Chen
- Department of Cardiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, No. 134, East Street, Gulou District, Fuzhou, 350001, Fujian, China
| | - Qunying Xi
- Center for Pulmonary Vascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, No. 12, Langshan Road, Shenzhen, 518057, Nanshan, China
| | - Zhihui Zhao
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China
| | - Qing Zhao
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China
| | - Tao Yang
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China
| | - Qixian Zeng
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China
| | - Xin Li
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China
| | - Zhihua Huang
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China
| | - Anqi Duan
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China
| | - Yijia Wang
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China
| | - Qin Luo
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China.
| | - Yansong Guo
- Department of Cardiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, No. 134, East Street, Gulou District, Fuzhou, 350001, Fujian, China.
| | - Zhihong Liu
- Center for Respiratory and Pulmonary Vascular Diseases, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Beijing, 100037, Xicheng, China.
| |
Collapse
|
22
|
Han Y, Sun Q, Chen W, Gao Y, Ye J, Chen Y, Wang T, Gao L, Liu Y, Yang Y. New advances of adiponectin in regulating obesity and related metabolic syndromes. J Pharm Anal 2024; 14:100913. [PMID: 38799237 PMCID: PMC11127227 DOI: 10.1016/j.jpha.2023.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/18/2023] [Accepted: 12/07/2023] [Indexed: 05/29/2024] Open
Abstract
Obesity and related metabolic syndromes have been recognized as important disease risks, in which the role of adipokines cannot be ignored. Adiponectin (ADP) is one of the key adipokines with various beneficial effects, including improving glucose and lipid metabolism, enhancing insulin sensitivity, reducing oxidative stress and inflammation, promoting ceramides degradation, and stimulating adipose tissue vascularity. Based on those, it can serve as a positive regulator in many metabolic syndromes, such as type 2 diabetes (T2D), cardiovascular diseases, non-alcoholic fatty liver disease (NAFLD), sarcopenia, neurodegenerative diseases, and certain cancers. Therefore, a promising therapeutic approach for treating various metabolic diseases may involve elevating ADP levels or activating ADP receptors. The modulation of ADP genes, multimerization, and secretion covers the main processes of ADP generation, providing a comprehensive orientation for the development of more appropriate therapeutic strategies. In order to have a deeper understanding of ADP, this paper will provide an all-encompassing review of ADP.
Collapse
Affiliation(s)
- Yanqi Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Qianwen Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Wei Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yue Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanmin Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Tingting Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Lili Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
23
|
Wu L, Ye S, Deng X, Fu Z, Li J, Yang C. Conjugated Linoleic Acid Ameliorates High Fat-Induced Insulin Resistance via Regulating Gut Microbiota-Host Metabolic and Immunomodulatory Interactions. Nutrients 2024; 16:1133. [PMID: 38674824 PMCID: PMC11053735 DOI: 10.3390/nu16081133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/20/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Interaction between gut microbiota, host immunity and metabolism has been suggested to crucially affect the development of insulin resistance (IR). This study aims to investigate how gut microbiota, inflammatory responses and metabolism in individuals with IR are affected by the supplementation of conjugated linoleic acid (CLA) and how this subsequently affects the pathophysiology of IR by using a high-fat diet-induced IR mouse model. Serum biochemical indices showed that 400 mg/kg body weight of CLA effectively attenuated hyperglycemia, hyperlipidemia, glucose intolerance and IR, while also promoting antioxidant capacities. Histomorphology, gene and protein expression analysis revealed that CLA reduced fat deposition and inflammation, and enhanced fatty acid oxidation, insulin signaling and glucose transport in adipose tissue or liver. Hepatic transcriptome analysis confirmed that CLA inhibited inflammatory signaling pathways and promoted insulin, PI3K-Akt and AMPK signaling pathways, as well as linoleic acid, arachidonic acid, arginine and proline metabolism. Gut microbiome analysis further revealed that these effects were highly associated with the enriched bacteria that showed positive correlation with the production of short-chain fatty acids (SCFAs), as well as the improved SCFAs production simultaneously. This study highlights the therapeutic actions of CLA on ameliorating IR via regulating microbiota-host metabolic and immunomodulatory interactions, which have important implications for IR control.
Collapse
Affiliation(s)
- Linjun Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China; (L.W.); (S.Y.); (X.D.)
| | - Shijie Ye
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China; (L.W.); (S.Y.); (X.D.)
| | - Xiangfei Deng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China; (L.W.); (S.Y.); (X.D.)
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China; (L.W.); (S.Y.); (X.D.)
| | - Jinjun Li
- Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
- Key Laboratory of Postharvest Preservation and Processing of Vegetables (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hangzhou 310021, China
- Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture and Rural Affairs, Hangzhou 310021, China
- Key Laboratory of Fruits and Vegetables Postharvest and Processing Technology Research of Zhejiang Province, Hangzhou 310021, China
| | - Chunlei Yang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China; (L.W.); (S.Y.); (X.D.)
| |
Collapse
|
24
|
Li Q, Byun J, Choi J, Park J, Lee J, Oh YK. Nanomodulator-Mediated Restructuring of Adipose Tissue Immune Microenvironments for Antiobesity Treatment. ACS NANO 2024; 18:9311-9330. [PMID: 38498418 DOI: 10.1021/acsnano.3c06001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
In obesity, the interactions between proinflammatory macrophages and adipocytes in white adipose tissues are known to play a crucial role in disease progression by providing inflammatory microenvironments. Here, we report that the functional nanoparticle-mediated modulation of crosstalk between adipocytes and macrophages can remodel adipocyte immune microenvironments. As a functional nanomodulator, we designed antivascular cell adhesion molecule (VCAM)-1 antibody-conjugated and amlexanox-loaded polydopamine nanoparticles (VAPN). Amlexanox was used as a model drug to increase energy expenditure. Compared to nanoparticles lacking antibody modification or amlexanox, VAPN showed significantly greater binding to VCAM-1-expressing adipocytes and lowered the interaction of adipocytes with macrophages. In high fat diet-fed mice, repeated subcutaneous administration of VAPN increased the populations of beige adipocytes and ameliorated inflammation in white adipose tissues. Moreover, the localized application of VAPN in vivo exerted a systemic metabolic effect and reduced metabolic disorders, including insulin tolerance and liver steatosis. These findings suggested that VAPN had potential to modulate the immune microenvironments of adipose tissues for the immunologic treatment of obesity. Although we used amlexanox as a model drug and anti-VCAM-1 antibody in VAPN, the concept of immune nanomodulators can be widely applied to the immunological treatment of obesity.
Collapse
Affiliation(s)
- Qiaoyun Li
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Junho Byun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaehyun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
25
|
Liu X, Chen X, Wang C, Song J, Xu J, Gao Z, Huang Y, Suo H. Mechanisms of probiotic modulation of ovarian sex hormone production and metabolism: a review. Food Funct 2024; 15:2860-2878. [PMID: 38433710 DOI: 10.1039/d3fo04345b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Sex hormones play a pivotal role in the growth and development of the skeletal, neurological, and reproductive systems. In women, the dysregulation of sex hormones can result in various health complications such as acne, hirsutism, and irregular menstruation. One of the most prevalent diseases associated with excess androgens is polycystic ovary syndrome with a hyperandrogenic phenotype. Probiotics have shown the potential to enhance the secretion of ovarian sex hormones. However, the underlying mechanism of action remains unclear. Furthermore, comprehensive reviews detailing how probiotics modulate ovarian sex hormones are scarce. This review seeks to shed light on the potential mechanisms through which probiotics influence the production of ovarian sex hormones. The role of probiotics across various biological axes, including the gut-ovarian, gut-brain-ovarian, gut-liver-ovarian, gut-pancreas-ovarian, and gut-fat-ovarian axes, with a focus on the direct impact of probiotics on the ovaries via the gut and their effects on brain gonadotropins is discussed. It is also proposed herein that probiotics can significantly influence the onset, progression, and complications of ovarian sex hormone abnormalities. In addition, this review provides a theoretical basis for the therapeutic application of probiotics in managing sex hormone-related health conditions.
Collapse
Affiliation(s)
- Xiao Liu
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
| | - Xiaoyong Chen
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Jiahui Xu
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
| | - Zhen Gao
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
| | - Yechuan Huang
- College of Bioengineering, Jingchu University of Technology, Jingmen 448000, P. R. China.
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| |
Collapse
|
26
|
Yang Y, Liu J, Kousteni S. Lipocalin 2-A bone-derived anorexigenic and β-cell promoting signal: From mice to humans. J Diabetes 2024; 16:e13504. [PMID: 38035773 PMCID: PMC10940901 DOI: 10.1111/1753-0407.13504] [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: 06/26/2023] [Revised: 10/16/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
The skeleton is traditionally known for its structural support, organ protection, movement, and maintenance of mineral homeostasis. Over the last 10 years, bone has emerged as an endocrine organ with diverse physiological functions. The two key molecules in this context are fibroblast growth factor 23 (FGF23), secreted by osteocytes, and osteocalcin, a hormone produced by osteoblasts. FGF23 affects mineral homeostasis through its actions on the kidneys, and osteocalcin has beneficial effects in improving glucose homeostasis, muscle function, brain development, cognition, and male fertility. In addition, another osteoblast-derived hormone, lipocalin 2 (LCN2) has emerged into the researchers' field of vision. In this review, we mainly focus on LCN2's role in appetite regulation and glucose metabolism and also briefly introduce its effects in other pathophysiological conditions, such as nonalcoholic fatty liver disease, sarcopenic obesity, and cancer-induced cachexia.
Collapse
Affiliation(s)
- Yuying Yang
- Department of Endocrine and Metabolic Diseases, Rui‐jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai Institute of Endocrine and Metabolic Diseases, and Shanghai Clinical Center for Endocrine and Metabolic DiseasesShanghaiChina
- Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Clinical Research Center for Metabolic Diseases, Shanghai National Center for Translational Medicine, Rui‐jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jianmin Liu
- Department of Endocrine and Metabolic Diseases, Rui‐jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai Institute of Endocrine and Metabolic Diseases, and Shanghai Clinical Center for Endocrine and Metabolic DiseasesShanghaiChina
- Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Clinical Research Center for Metabolic Diseases, Shanghai National Center for Translational Medicine, Rui‐jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Stavroula Kousteni
- Department of Physiology and Cellular BiophysicsColumbia University Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
27
|
Ali M, Kumari T, Gupta A, Akhtar S, Verma RD, Ghosh JK. Identification of a 10-mer peptide from the death domain of MyD88 which attenuates inflammation and insulin resistance and improves glucose metabolism. Biochem J 2024; 481:191-218. [PMID: 38224573 DOI: 10.1042/bcj20230369] [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: 08/26/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/17/2024]
Abstract
Insulin resistance (IR) is the key pathophysiological cause of type 2 diabetes, and inflammation has been implicated in it. The death domain (DD) of the adaptor protein, MyD88 plays a crucial role in the transduction of TLR4-associated inflammatory signal. Herein, we have identified a 10-residue peptide (M10), from the DD of MyD88 which seems to be involved in Myddosome formation. We hypothesized that M10 could inhibit MyD88-dependent TLR4-signaling and might have effects on inflammation-associated IR. Intriguingly, 10-mer M10 showed oligomeric nature and reversible self-assembly property indicating the peptide's ability to recognize its own amino acid sequence. M10 inhibited LPS-induced nuclear translocation of NF-κB in L6 myotubes and also reduced LPS-induced IL-6 and TNF-α production in peritoneal macrophages of BALB/c mice. Remarkably, M10 inhibited IL-6 and TNF-α secretion in diabetic, db/db mice. Notably, M10 abrogated IR in insulin-resistant L6 myotubes, which was associated with an increase in glucose uptake and a decrease in Ser307-phosphorylation of IRS1, TNF-α-induced JNK activation and nuclear translocation of NF-κB in these cells. Alternate day dosing with M10 (10 and 20 mg/kg) for 30 days in db/db mice significantly lowered blood glucose and improved glucose intolerance after loading, 3.0 g/kg glucose orally. Furthermore, M10 increased insulin and adiponectin secretion in db/db mice. M10-induced glucose uptake in L6 myotubes involved the activation of PI3K/AKT/GLUT4 pathways. A scrambled M10-analog was mostly inactive. Overall, the results show the identification of a 10-mer peptide from the DD of MyD88 with anti-inflammatory and anti-diabetic properties, suggesting that targeting of TLR4-inflammatory pathway, could lead to the discovery of molecules against IR and diabetes.
Collapse
Affiliation(s)
- Mehmood Ali
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 India
| | - Tripti Kumari
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
| | - Arvind Gupta
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 India
| | - Sariyah Akhtar
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
| | - Rahul Dev Verma
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 India
| | - Jimut Kanti Ghosh
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 India
| |
Collapse
|
28
|
McGregor ER, Lasky DJ, Rippentrop OJ, Clark JP, Wright SLG, Jones MV, Anderson RM. Reversal of neuronal tau pathology, metabolic dysfunction, and electrophysiological defects via adiponectin pathway-dependent AMPK activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.07.579204. [PMID: 38370802 PMCID: PMC10871331 DOI: 10.1101/2024.02.07.579204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Changes in brain mitochondrial metabolism are coincident with functional decline; however, direct links between the two have not been established. Here, we show that mitochondrial targeting via the adiponectin receptor activator AdipoRon (AR) clears neurofibrillary tangles (NFTs) and rescues neuronal tauopathy-associated defects. AR reduced levels of phospho-tau and lowered NFT burden by a mechanism involving the energy-sensing kinase AMPK and the growth-sensing kinase GSK3b. The transcriptional response to AR included broad metabolic and functional pathways. Induction of lysosomal pathways involved activation of LC3 and p62, and restoration of neuronal outgrowth required the stress-responsive kinase JNK. Negative consequences of NFTs on mitochondrial activity, ATP production, and lipid stores were corrected. Defects in electrophysiological measures (e.g., resting potential, resistance, spiking profiles) were also corrected. These findings reveal a network linking mitochondrial function, cellular maintenance processes, and electrical aspects of neuronal function that can be targeted via adiponectin receptor activation.
Collapse
Affiliation(s)
- Eric R McGregor
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Danny J Lasky
- Department. of Neuroscience, Univ. of Wisconsin-Madison, Madison, WI
| | | | - Josef P Clark
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI
| | | | - Mathew V Jones
- Department. of Neuroscience, Univ. of Wisconsin-Madison, Madison, WI
| | - Rozalyn M Anderson
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
- GRECC William S. Middleton Memorial Veterans Hospital, Madison, WI
| |
Collapse
|
29
|
Moreno LG, César NR, Melo DS, Figueiró MTO, Dos Santos EC, Evangelista-Silva PH, de Sousa Santos C, Costa KB, Rocha-Vieira E, Dias-Peixoto MF, Castro Magalhães FD, Esteves EA. A MUFA/carotenoid-rich oil ameliorated insulin resistance by improving inflammation and oxidative stress in obese rats. Mol Cell Endocrinol 2024; 581:112110. [PMID: 37981187 DOI: 10.1016/j.mce.2023.112110] [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: 06/21/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
Obesity is associated with low-grade inflammation and oxidative stress, leading to insulin resistance and type II diabetes. Caryocar brasiliense pulp oil (pequi oil - PO) is rich in oleic acid and carotenoids and positively implicated in regulating inflammation and oxidative stress. This study investigated PO's antioxidant and anti-inflammatory effects in a diet-induced obesity model. Male Wistar rats were allocated into three experimental groups: Control (CD), Western Diet (WD), and Western Diet, with 27% of lard switched by PO (WDP). Metabolic, inflammatory, and oxidative stress biomarkers were evaluated after 12 weeks of diet protocols in liver and adipose tissue. WDP rats gained less body mass and epididymal fat, had less hepatic fat infiltration, and were more glucose-tolerant and insulin-sensitive than WD (p < 0.05). In the liver, the WDP group had the highest non-enzymatic antioxidant capacity, SOD and GPx activities, CAT, SOD II, and HSP72 expression compared to WD (p < 0.05). Adipose tissue IL-6 and TNF were reduced, and IL-10 was increased in WDP compared to WD (p < 0.05). Our data suggest that the partial replacement of lard by PO in a Western diet prevented visceral fat accumulation and contributed to reducing inflammation in adipose tissue and liver oxidative stress, improving obesity-related insulin resistance.
Collapse
Affiliation(s)
- Lauane Gomes Moreno
- Programa de Pós-Graduação Em Ciências da Saúde, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri - UFVJM, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil.
| | - Nayara Rayane César
- Programa de Pós-graduação Multicêntrico Em Ciências Fisiológicas, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil.
| | - Dirceu Sousa Melo
- Instituto de Ciências Naturais, Departamento de Biologia, Universidade Federal de Lavras - UFLA, Aquenta Sol, Lavras, MG, 37200-900, Brazil.
| | - Maria Thereza Otoni Figueiró
- Programa de Pós-Graduação Em Ciências da Saúde, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri - UFVJM, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil.
| | - Edivânia Cordeiro Dos Santos
- Programa de Pós-graduação Multicêntrico Em Ciências Fisiológicas, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil.
| | | | - Carina de Sousa Santos
- Faculdade de Ciências da Saúde, Curso de Nutrição, Universidade Federal de Grande Dourados - UFGD, Dourados, Brazil.
| | - Karine Beatriz Costa
- Programa de Pós-graduação Em Ciências Aplicadas à Saúde - PPgCAS, Universidade Federal de Juiz de Fora - UFJF, Governador Valadares, MG, 35010-180, Brazil.
| | - Etel Rocha-Vieira
- Programa de Pós-Graduação Em Ciências da Saúde, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri - UFVJM, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil; Programa de Pós-graduação Multicêntrico Em Ciências Fisiológicas, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil.
| | - Marco Fabrício Dias-Peixoto
- Programa de Pós-Graduação Em Ciências da Saúde, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri - UFVJM, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil; Programa de Pós-graduação Multicêntrico Em Ciências Fisiológicas, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil.
| | - Flávio de Castro Magalhães
- Programa de Pós-Graduação Em Ciências da Saúde, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri - UFVJM, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil; Programa de Pós-graduação Multicêntrico Em Ciências Fisiológicas, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil.
| | - Elizabethe Adriana Esteves
- Programa de Pós-Graduação Em Ciências da Saúde, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri - UFVJM, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil; Programa de Pós-graduação Multicêntrico Em Ciências Fisiológicas, Universidade Federal Dos Vales Do Jequitinhonha e Mucuri, Rodovia MGT 367 - Km 583. N. 5000, Alto da Jacuba, Diamantina, MG, 39100-000, Brazil.
| |
Collapse
|
30
|
Boachie J, Zammit V, Saravanan P, Adaikalakoteswari A. Metformin Inefficiency to Lower Lipids in Vitamin B12 Deficient HepG2 Cells Is Alleviated via Adiponectin-AMPK Axis. Nutrients 2023; 15:5046. [PMID: 38140305 PMCID: PMC10745523 DOI: 10.3390/nu15245046] [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: 10/23/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Background: Prolonged metformin treatment decreases vitamin B12 (B12) levels, whereas low B12 is associated with dyslipidaemia. Some studies have reported that metformin has no effect on intrahepatic triglyceride (TG) levels. Although AMP-activated protein kinase (AMPK) activation via adiponectin lowers hepatic TG content, its role in B12 deficiency and metformin has not been explored. We investigated whether low B12 impairs the beneficial effect of metformin on hepatic lipid metabolism via the AMPK-adiponectin axis. Methods: HepG2 was cultured using custom-made B12-deficient Eagle's Minimal Essential Medium (EMEM) in different B12-medium concentrations, followed by a 24-h metformin/adiponectin treatment. Gene and protein expressions and total intracellular TG were measured, and radiochemical analysis of TG synthesis and seahorse mitochondria stress assay were undertaken. Results: With low B12, total intracellular TG and synthesized radiolabelled TG were increased. Regulators of lipogenesis, cholesterol and genes regulating fatty acids (FAs; TG; and cholesterol biosynthesis were increased. FA oxidation (FAO) and mitochondrial function were decreased, with decreased pAMPKα and pACC levels. Following metformin treatment in hepatocytes with low B12, the gene and protein expression of the above targets were not alleviated. However, in the presence of adiponectin, intrahepatic lipid levels with low B12 decreased via upregulated pAMPKα and pACC levels. Again, combined adiponectin and metformin treatment ameliorated the low B12 effect and resulted in increased pAMPKα and pACC, with a subsequent reduction in lipogenesis, increased FAO and mitochondrion function. Conclusions: Adiponectin co-administration with metformin induced a higher intrahepatic lipid-lowering effect. Overall, we emphasize the potential therapeutic implications for hepatic AMPK activation via adiponectin for a clinical condition associated with B12 deficiency and metformin treatment.
Collapse
Affiliation(s)
- Joseph Boachie
- Division of Metabolic and Vascular Health, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital-Walsgrave Campus, Coventry CV2 2DX, UK; (J.B.); (V.Z.); (P.S.)
| | - Victor Zammit
- Division of Metabolic and Vascular Health, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital-Walsgrave Campus, Coventry CV2 2DX, UK; (J.B.); (V.Z.); (P.S.)
| | - Ponnusamy Saravanan
- Division of Metabolic and Vascular Health, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital-Walsgrave Campus, Coventry CV2 2DX, UK; (J.B.); (V.Z.); (P.S.)
- Diabetes Centre, George Eliot Hospital NHS Trust, College Street, Nuneaton CV10 7DJ, UK
- Populations, Evidence and Technologies, Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7HL, UK
| | - Antonysunil Adaikalakoteswari
- Division of Metabolic and Vascular Health, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital-Walsgrave Campus, Coventry CV2 2DX, UK; (J.B.); (V.Z.); (P.S.)
- Department of Bioscience, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK
| |
Collapse
|
31
|
Zheng W, Yang J, Zhang Q, Cheng M, Shaukat H, Qin H. Sesamol Alleviates High-Fat Diet-Induced Hepatic Insulin Resistance in C57BL/6 J Mice Through AMPK Activation Mediated by Adipose Adiponectin. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2023; 78:720-727. [PMID: 37775709 DOI: 10.1007/s11130-023-01108-z] [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] [Accepted: 09/22/2023] [Indexed: 10/01/2023]
Abstract
Sesamol is the major bioactive constituent isolated from sesame seeds and has a variety of bioactivities. However, its role and mechanism in liver insulin resistance remain unknown. The current study was designed to investigate the underlying adipose-liver crosstalk mechanism of sesamol ameliorating hepatic insulin sensitivity. The therapeutic effect of sesamol was evaluated in high-fat diet (HFD)-fed C57BL/6 J mice (100 mg/kg for 8 weeks, XYGW-2021-75) and the mechanism was further explored in HepG2 cells with/without adiponectin and adenosine 5 '-monophosphate-activated protein kinase (AMPK) inhibitor administration. Our in vivo data showed that sesamol reduced hepatic insulin resistance in HFD-induced mice with obesity by modulating protein expression levels of glycogen synthase (GS), phosphoenolpyruvate carboxykinase (PEPCK) and protein kinase B (AKT). Moreover, sesamol not only increased the serum and adipose tissue adiponectin concentrations but also activated the phosphorylation of AMPK in the liver. Furthermore, in vitro studies using recombinant human adiponectin and an AMPK inhibitor revealed that adiponectin and sesamol have a synergic impact on increasing glycogenesis and reducing gluconeogenesis, of which the effects could be attenuated by the AMPK inhibitor. Taken together, our results suggested that sesamol stimulated adiponectin secretion from adipocytes, whereby exhibited a co-effect on activating the downstream signal of hepatic AMPK, resulting in the alleviation of hepatic insulin resistance. The novel findings of sesamol on hepatic effects provides prospective therapeutic approaches to treat insulin resistance.
Collapse
Affiliation(s)
- Wenya Zheng
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, China
| | - Jinxin Yang
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, China
| | - Quanquan Zhang
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, China
| | - Minghui Cheng
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, China
| | - Horia Shaukat
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, China
| | - Hong Qin
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
32
|
Barbalho SM, Méndez-Sánchez N, Fornari Laurindo L. AdipoRon and ADP355, adiponectin receptor agonists, in Metabolic-associated Fatty Liver Disease (MAFLD) and Nonalcoholic Steatohepatitis (NASH): A systematic review. Biochem Pharmacol 2023; 218:115871. [PMID: 37866803 DOI: 10.1016/j.bcp.2023.115871] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
Adiponectin replacement therapy holds the potential to benefit numerous human diseases, and ongoing research applies particular interest in how adiponectin acts against Metabolic-associated Fatty Liver Disease (MAFLD) and Nonalcoholic Steatohepatitis (NASH). However, the pharmacological limitations of the intact protein have prompted a focus on alternative options, specifically peptidic and small molecule agonists targeting the adiponectin receptor. AdipoRon is an extensively researched non-peptidic drug candidate in adiponectin replacement therapy. In turn, ADP355 is an adiponectin-based active short peptide. They have garnered significant attention due to their potential as substitutes for adiponectin. Researchers have studied AdipoRon's and ADP355's efficacy and therapeutic applications in various disease conditions. However, the effects of AdipoRon and ADP355 against NAFLD and NASH models advanced more, and no systematic review explored this area before. This systematic review was conceived to address the deficiency mentioned above and consider the lack of clinical evidence. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were utilized. To assess the risk of bias in systematic review, The Joanna Briggs Institute (JBI) Critical Appraisal Checklist was employed. Results from pre-clinical evidence show that AdipoRon and ADP355 represent promising effects in NAFLD and NASH-related models, including reducing hepatic steatosis, modulating inflammation, improving insulin sensitivity, enhancing mitochondrial function, and protecting against liver fibrosis. While AdipoRon and ADP355 exhibit promise in pre-clinical studies and experimental models, additional clinical trials are necessary to assess their effectiveness, safety, and potential translational therapeutic potential uses in NAFLD and NASH human cases.
Collapse
Affiliation(s)
- Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), São Paulo, Brazil.
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico; Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), São Paulo, Brazil; Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília, São Paulo, Brazil
| |
Collapse
|
33
|
Xia W, Li S, Li L, Zhang S, Wang X, Ding W, Ding L, Zhang X, Wang Z. Role of anthraquinones in combating insulin resistance. Front Pharmacol 2023; 14:1275430. [PMID: 38053837 PMCID: PMC10694622 DOI: 10.3389/fphar.2023.1275430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
Insulin resistance presents a formidable public health challenge that is intricately linked to the onset and progression of various chronic ailments, including diabetes, cardiovascular disease, hypertension, metabolic syndrome, nonalcoholic fatty liver disease, and cancer. Effectively addressing insulin resistance is paramount in preventing and managing these metabolic disorders. Natural herbal remedies show promise in combating insulin resistance, with anthraquinone extracts garnering attention for their role in enhancing insulin sensitivity and treating diabetes. Anthraquinones are believed to ameliorate insulin resistance through diverse pathways, encompassing activation of the AMP-activated protein kinase (AMPK) signaling pathway, restoration of insulin signal transduction, attenuation of inflammatory pathways, and modulation of gut microbiota. This comprehensive review aims to consolidate the potential anthraquinone compounds that exert beneficial effects on insulin resistance, elucidating the underlying mechanisms responsible for their therapeutic impact. The evidence discussed in this review points toward the potential utilization of anthraquinones as a promising therapeutic strategy to combat insulin resistance and its associated metabolic diseases.
Collapse
Affiliation(s)
- Wanru Xia
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shuqian Li
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - LinZehao Li
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shibo Zhang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaolei Wang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wenyu Ding
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Lina Ding
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiandang Zhang
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhibin Wang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
34
|
Lai HC, Chen PH, Tang CH, Chen LW. Dipeptidyl Peptidase 4 Stimulation Induces Adipogenesis-Related Gene Expression of Adipose Stromal Cells. Int J Mol Sci 2023; 24:16101. [PMID: 38003291 PMCID: PMC10671339 DOI: 10.3390/ijms242216101] [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: 09/17/2023] [Revised: 11/01/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Adipogenesis has emerged as a new therapeutic target for regulating metabolism and achieving anti-inflammatory and anti-atherosclerotic effects via the release of adiponectin. However, at present, the effects and mechanism of action of dipeptidyl peptidase 4 (DPP4) stimulation on adiponectin production and adipogenesis have not been clarified. Here, we investigated the effects of DPP4 stimulation with monocyte chemoattractant protein-1 (MCP-1) on platelet-derived growth factor receptor alpha (PDGFRα) expression in adipose tissue and blood adiponectin levels. Stromal vascular fractions (SVFs) purified from human subcutaneous adipose tissue and inguinal adipose tissue of obese and diabetic (Leprdb/db) mice were treated with 50 ng of MCP-1 and plasma from control (Lepr+/+) mice supplemented with 10 ng or 50 ng of MCP-1. Treatment of SVFs from human subcutaneous adipose tissues with 50 ng of MCP-1 significantly increased AdipoQ, DPP4, peroxisome proliferator-activated receptor gamma (PPARγ), fatty-acid-binding protein (FABP4), and SERBF1 mRNA expression. MCP-1-supplemented plasma increased adiponectin, CCAAT-Enhancer-binding protein alpha (C/EBPα), DPP4, IL-33, and PDGFRα mRNA expression and adiponectin and DPP4 protein expression, while decreasing the expression of IL-10 mRNA in SVFs compared with the levels in the plasma treatment group. MCP-1-supplemented plasma was shown to increase PPARγ, PPARγ2, adiponectin, DPP4, and FABP4 and decrease IL-10 mRNA expression in PDGFRα cells from adipose tissue. Meanwhile, MCP-1-supplemented plasma increased MCP-1, PDGFRα, TNFα, adiponectin, and IL-1β and decreased IL-10 and FOXP3 mRNA expression in DPP4 cells. Moreover, the injection of MCP-1-supplemented plasma into adipose tissue increased the proportion of DPP4+ cells among PDGFRα+ cells from adipose tissue and plasma adiponectin levels of Leprdb/db mice compared with the levels in the plasma injection group. Our results demonstrate that DPP4+ cells are important adipose progenitor cells. Stimulation of DPP4 with MCP-1 increases adipogenesis-related gene expression and the population of DPP4+ cells among PDGFRα+ cells in SVFs and blood adiponectin levels. DPP4 stimulation could be a novel therapy to increase local adipogenesis and systemic adiponectin levels.
Collapse
Affiliation(s)
- Hsiao-Chi Lai
- Department of Surgery, Kaohsiung Veterans General Hospital, No. 386, Ta-Chung 1st Road, Kaohsiung 813, Taiwan
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei 112, Taiwan
| | - Pei-Hsuan Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, No. 386, Ta-Chung 1st Road, Kaohsiung 813, Taiwan
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei 112, Taiwan
| | - Chia-Hua Tang
- Department of Surgery, Kaohsiung Veterans General Hospital, No. 386, Ta-Chung 1st Road, Kaohsiung 813, Taiwan
| | - Lee-Wei Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, No. 386, Ta-Chung 1st Road, Kaohsiung 813, Taiwan
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei 112, Taiwan
- Department of Biological Sciences, National Sun Yat-Sen University, No. 70, Lien-Hai Road, Kaohsiung 804, Taiwan
| |
Collapse
|
35
|
Domingues I, Leclercq IA, Beloqui A. Nonalcoholic fatty liver disease: Current therapies and future perspectives in drug delivery. J Control Release 2023; 363:415-434. [PMID: 37769817 DOI: 10.1016/j.jconrel.2023.09.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 08/27/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects approximately 25% of the adult population worldwide. This pathology can progress into end-stage liver disease with life-threatening complications, and yet no pharmacologic therapy has been approved. NAFLD is commonly characterized by excessive fat accumulation in the liver and is in closely associated with insulin resistance and metabolic disorders, which suggests that NAFLD is the hepatic manifestation of metabolic syndrome. Regarding treatment options, the current validated strategy relies on lifestyle modifications (exercise and diet restrictions). Although there are no approved drug-based treatments, several clinical trials are ongoing. Novel targets are being discovered, and the repurposing of drugs that show promising effects in NAFLD is starting to gain more interest. The field of nanotechnology has been growing at an increasing rate, with new and more efficient drug delivery strategies being developed for NAFLD treatment. Nanocarriers can easily encapsulate drugs that need to be better protected from the organism to exert their effect or that need help at reaching their target, thereby helping achieve a better bioavailability. Drug delivery systems can also be designed to target the site of the disease, in this case, the liver. In this review, we focus on the current knowledge of NAFLD pathology, the targets being considered for clinical trials, and the current guidelines and ongoing clinical trials, with a specific focus on potential oral treatments for NAFLD using promising drug delivery strategies.
Collapse
Affiliation(s)
- Inês Domingues
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials Group, Avenue Emmanuel Mounier 73, 1200 Brussels, Belgium
| | - Isabelle A Leclercq
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research, Laboratory of Hepato-Gastroenterology, Avenue Emmanuel Mounier 53, 1200 Brussels, Belgium.
| | - Ana Beloqui
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials Group, Avenue Emmanuel Mounier 73, 1200 Brussels, Belgium; WEL Research Institute, Avenue Pasteur, 6, 1300 Wavre, Belgium.
| |
Collapse
|
36
|
Basu A, Hooyman A, Richardson LA, Alman AC, Snell-Bergeon JK. Longitudinal Associations of Dietary Fiber Intake with Glycated Hemoglobin and Estimated Insulin Sensitivity in Adults with and without Type 1 Diabetes. Nutrients 2023; 15:4620. [PMID: 37960272 PMCID: PMC10648902 DOI: 10.3390/nu15214620] [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: 10/13/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Dietary fiber, an essential bioactive compound in plant-based diets, is of public health concern based on habitual low intakes in the general population. Not much data are available on how habitual dietary fiber is associated with glycemic control in type 1 diabetes (T1D) as well as in prediabetes and normoglycemic adults. To address this gap, we conducted a six-year longitudinal analysis of an original cohort in adults with and without T1D (n = 1255; T1D: n = 563; non-diabetes mellitus (non-DM): n = 692). Dietary data were collected from a validated food frequency questionnaire, biochemical measures were obtained after an overnight fast, and anthropometric measurements were collected at baseline as well as after three and six years for the follow-up study. Glycated hemoglobin (HbA1c) and estimated insulin sensitivity (eIS) were the main outcomes examined. In adjusted analyses, dietary fiber intake was inversely associated with HbA1c in a minimally adjusted model, but it was positively associated with eIS in a model involving all relevant covariates in non-DM adults. These associations were not significant in the T1D group. Furthermore, when examined by HbA1c cut-offs for glycemic control, an inverse association with dietary fiber was only observed in adults with prediabetes (all p < 0.05). At a six-year mean (±SD) dietary fiber intake of 17.4 ± 8.8 g for non-DM and 17.0 ± 8.2 g for the T1D group, protective associations against poor glycemic control were observed in those without diabetes and in prediabetes.
Collapse
Affiliation(s)
- Arpita Basu
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
| | - Andrew Hooyman
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
- School of Biological Health Systems Engineering, Arizona State University, Tempe, AZ 85281, USA
| | - Leigh Ann Richardson
- Department of Epidemiology and Biostatistics, School of Public Health, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
| | - Amy C. Alman
- College of Public Health, University of South Florida, Tampa, FL 33620, USA;
| | - Janet K. Snell-Bergeon
- Barbara Davis Center for Diabetes, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA;
| |
Collapse
|
37
|
Bo B, Guo A, Kaila SJ, Hao Z, Zhang H, Wei J, Yao Y. Elucidating the primary mechanisms of high-intensity interval training for improved cardiac fitness in obesity. Front Physiol 2023; 14:1170324. [PMID: 37608837 PMCID: PMC10441243 DOI: 10.3389/fphys.2023.1170324] [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: 02/20/2023] [Accepted: 07/31/2023] [Indexed: 08/24/2023] Open
Abstract
Obesity is a global and rising multifactorial pandemic associated with the emergence of several comorbidities that are risk factors for malignant cardiac remodeling and disease. High-intensity interval training (HIIT) has gained considerable attention due to its favorable outcomes of cardiometabolic health in individuals with overweight or obese. The primary aim of this review is to discuss the fundamental processes through which HIIT improves cardiac impairment in individuals with obesity to develop viable treatments for obesity management. In this review, a multiple database search and collection were conducted from the earliest record to January 2013 for studies included the qualitative component of HIIT intervention in humans and animals with overweight/obesity related to cardiac remodeling and fitness. We attempt to integrate the main mechanisms of HIIT in cardiac remolding improvement in obesity into an overall sequential hypothesis. This work focus on the ameliorative effects of HIIT on obesity-induced cardiac remodeling with respect to potential and pleiotropic mechanisms, including adipose distribution, energy metabolism, inflammatory response, insulin resistance, and related risk profiles in obesity. In conclusion, HIIT has been shown to reduce obesity-induced risks of cardiac remodeling, but the long-term effects of HIIT on obesity-induced cardiac injury and disease are presently unknown. Collective understanding highlights numerous specific research that are needed before the safety and effectiveness of HIIT can be confirmed and widely adopted in patient with obesity.
Collapse
Affiliation(s)
- Bing Bo
- Department of Kinesiology, School of Physical Education, Henan University, Kaifeng, China
- Sports Reform and Development Research Center, School of Physical Education, Henan University, Kaifeng, China
| | - Aijing Guo
- Department of Kinesiology, School of Physical Education, Henan University, Kaifeng, China
| | - Severa Jafeth Kaila
- Department of Kinesiology, School of Physical Education, Henan University, Kaifeng, China
| | - Zhe Hao
- Department of Kinesiology, School of Physical Education, Henan University, Kaifeng, China
| | - Huiqing Zhang
- Sports Reform and Development Research Center, School of Physical Education, Henan University, Kaifeng, China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, China
| | - Yuan Yao
- Sports Reform and Development Research Center, School of Physical Education, Henan University, Kaifeng, China
| |
Collapse
|
38
|
Wilson RA, Arivazhagan L, Ruiz HH, Zhou B, Qian K, Manigrasso MB, Bernadin R, Mangar K, Shekhtman A, Li H, Ramasamy R, Schmidt AM. Pharmacological antagonism of receptor for advanced glycation end products signaling promotes thermogenesis, healthful body mass and composition, and metabolism in mice. Obesity (Silver Spring) 2023; 31:1825-1843. [PMID: 37231626 PMCID: PMC10790363 DOI: 10.1002/oby.23774] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/14/2023] [Accepted: 02/28/2023] [Indexed: 05/27/2023]
Abstract
OBJECTIVE Optimal body mass and composition as well as metabolic fitness require tightly regulated and interconnected mechanisms across tissues. Disturbances in these regulatory networks tip the balance between metabolic health versus overweight and obesity and their complications. The authors previously demonstrated roles for the receptor for advanced glycation end products (RAGE) in obesity, as global- or adipocyte-specific deletion of Ager (the gene encoding RAGE) protected mice from high-fat diet-induced obesity and metabolic dysfunction. METHODS To explore translational strategies evoked by these observations, a small molecule antagonist of RAGE signaling, RAGE229, was administered to lean mice and mice with obesity undergoing diet-induced weight loss. Body mass and composition and whole body and adipose tissue metabolism were examined. RESULTS This study demonstrates that antagonism of RAGE signaling reduced body mass and adiposity and improved glucose, insulin, and lipid metabolism in lean male and female mice and in male mice with obesity undergoing weight loss. In adipose tissue and in human and mouse adipocytes, RAGE229 enhanced phosphorylation of protein kinase A substrates, which augmented lipolysis, mitochondrial function, and thermogenic programs. CONCLUSIONS Pharmacological antagonism of RAGE signaling is a potent strategy to optimize healthful body mass and composition and metabolic fitness.
Collapse
Affiliation(s)
- Robin A. Wilson
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Lakshmi Arivazhagan
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Henry H. Ruiz
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Boyan Zhou
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Kun Qian
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Michaele B. Manigrasso
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Rollanda Bernadin
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Kaamashri Mangar
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Alexander Shekhtman
- Department of Chemistry, State University of New York, Albany, New York, USA
| | - Huilin Li
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
39
|
Cao R, Tian H, Zhang Y, Liu G, Xu H, Rao G, Tian Y, Fu X. Signaling pathways and intervention for therapy of type 2 diabetes mellitus. MedComm (Beijing) 2023; 4:e283. [PMID: 37303813 PMCID: PMC10248034 DOI: 10.1002/mco2.283] [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: 01/06/2023] [Revised: 04/18/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) represents one of the fastest growing epidemic metabolic disorders worldwide and is a strong contributor for a broad range of comorbidities, including vascular, visual, neurological, kidney, and liver diseases. Moreover, recent data suggest a mutual interplay between T2DM and Corona Virus Disease 2019 (COVID-19). T2DM is characterized by insulin resistance (IR) and pancreatic β cell dysfunction. Pioneering discoveries throughout the past few decades have established notable links between signaling pathways and T2DM pathogenesis and therapy. Importantly, a number of signaling pathways substantially control the advancement of core pathological changes in T2DM, including IR and β cell dysfunction, as well as additional pathogenic disturbances. Accordingly, an improved understanding of these signaling pathways sheds light on tractable targets and strategies for developing and repurposing critical therapies to treat T2DM and its complications. In this review, we provide a brief overview of the history of T2DM and signaling pathways, and offer a systematic update on the role and mechanism of key signaling pathways underlying the onset, development, and progression of T2DM. In this content, we also summarize current therapeutic drugs/agents associated with signaling pathways for the treatment of T2DM and its complications, and discuss some implications and directions to the future of this field.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Huimin Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yu Zhang
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Geng Liu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Haixia Xu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Guocheng Rao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yan Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Xianghui Fu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
40
|
Akingbesote ND, Leitner BP, Jovin DG, Desrouleaux R, Owusu D, Zhu W, Li Z, Pollak MN, Perry RJ. Gene and protein expression and metabolic flux analysis reveals metabolic scaling in liver ex vivo and in vivo. eLife 2023; 12:e78335. [PMID: 37219930 PMCID: PMC10205083 DOI: 10.7554/elife.78335] [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: 03/03/2022] [Accepted: 05/08/2023] [Indexed: 05/24/2023] Open
Abstract
Metabolic scaling, the inverse correlation of metabolic rates to body mass, has been appreciated for more than 80 years. Studies of metabolic scaling have largely been restricted to mathematical modeling of caloric intake and oxygen consumption, and mostly rely on computational modeling. The possibility that other metabolic processes scale with body size has not been comprehensively studied. To address this gap in knowledge, we employed a systems approach including transcriptomics, proteomics, and measurement of in vitro and in vivo metabolic fluxes. Gene expression in livers of five species spanning a 30,000-fold range in mass revealed differential expression according to body mass of genes related to cytosolic and mitochondrial metabolic processes, and to detoxication of oxidative damage. To determine whether flux through key metabolic pathways is ordered inversely to body size, we applied stable isotope tracer methodology to study multiple cellular compartments, tissues, and species. Comparing C57BL/6 J mice with Sprague-Dawley rats, we demonstrate that while ordering of metabolic fluxes is not observed in in vitro cell-autonomous settings, it is present in liver slices and in vivo. Together, these data reveal that metabolic scaling extends beyond oxygen consumption to other aspects of metabolism, and is regulated at the level of gene and protein expression, enzyme activity, and substrate supply.
Collapse
Affiliation(s)
- Ngozi D Akingbesote
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Brooks P Leitner
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Daniel G Jovin
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Reina Desrouleaux
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Comparative Medicine, Yale UniversityNew HavenUnited States
| | - Dennis Owusu
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Wanling Zhu
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Zongyu Li
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Michael N Pollak
- Lady Davis Institute for Medical Research, Jewish General HospitalMontrealCanada
- Department of Oncology, McGill UniversityMontrealCanada
| | - Rachel J Perry
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| |
Collapse
|
41
|
Würfel M, Blüher M, Stumvoll M, Ebert T, Kovacs P, Tönjes A, Breitfeld J. Adipokines as Clinically Relevant Therapeutic Targets in Obesity. Biomedicines 2023; 11:biomedicines11051427. [PMID: 37239098 DOI: 10.3390/biomedicines11051427] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Adipokines provide an outstanding role in the comprehensive etiology of obesity and may link adipose tissue dysfunction to further metabolic and cardiovascular complications. Although several adipokines have been identified in terms of their physiological roles, many regulatory circuits remain unclear and translation from experimental studies to clinical applications has yet to occur. Nevertheless, due to their complex metabolic properties, adipokines offer immense potential for their use both as obesity-associated biomarkers and as relevant treatment strategies for overweight, obesity and metabolic comorbidities. To provide an overview of the current clinical use of adipokines, this review summarizes clinical studies investigating the potential of various adipokines with respect to diagnostic and therapeutic treatment strategies for obesity and linked metabolic disorders. Furthermore, an overview of adipokines, for which a potential for clinical use has been demonstrated in experimental studies to date, will be presented. In particular, promising data revealed that fibroblast growth factor (FGF)-19, FGF-21 and leptin offer great potential for future clinical application in the treatment of obesity and related comorbidities. Based on data from animal studies or other clinical applications in addition to obesity, adipokines including adiponectin, vaspin, resistin, chemerin, visfatin, bone morphogenetic protein 7 (BMP-7) and tumor necrosis factor alpha (TNF-α) provide potential for human clinical application.
Collapse
Affiliation(s)
- Marleen Würfel
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Liebigstr. 18, 04103 Leipzig, Germany
| | - Matthias Blüher
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Liebigstr. 18, 04103 Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Center Munich at the University of Leipzig and the University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Michael Stumvoll
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Liebigstr. 18, 04103 Leipzig, Germany
| | - Thomas Ebert
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Liebigstr. 18, 04103 Leipzig, Germany
| | - Peter Kovacs
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Liebigstr. 18, 04103 Leipzig, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Anke Tönjes
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Liebigstr. 18, 04103 Leipzig, Germany
| | - Jana Breitfeld
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Liebigstr. 18, 04103 Leipzig, Germany
| |
Collapse
|
42
|
Arjunan A, Song J. Pharmacological and physiological roles of adipokines and myokines in metabolic-related dementia. Biomed Pharmacother 2023; 163:114847. [PMID: 37150030 DOI: 10.1016/j.biopha.2023.114847] [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: 03/21/2023] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 05/09/2023] Open
Abstract
Dementia is a detrimental neuropathologic condition with considerable physical, mental, social, and financial impact on patients and society. Patients with metabolic syndrome (MetS), a group of diseases that occur in tandem and increase the risk of neurologic diseases, have a higher risk of dementia. The ratio between muscle and adipose tissue is crucial in MetS, as these contain many hormones, including myokines and adipokines, which are involved in crosstalk and local paracrine/autocrine interactions. Evidence suggests that abnormal adipokine and myokine synthesis and release may be implicated in various MetS, such as atherosclerosis, diabetic mellitus (DM), and dyslipidemia, but their precise role is unclear. Here we review the literature on adipokine and myokine involvement in MetS-induced dementia via glucose and insulin homeostasis regulation, neuroinflammation, vascular dysfunction, emotional changes, and cognitive function.
Collapse
Affiliation(s)
- Archana Arjunan
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
| |
Collapse
|
43
|
Liao LP, Wu L, Yang Y. The relationship between triglyceride/high-density lipoprotein cholesterol ratio and coronary microvascular disease. BMC Cardiovasc Disord 2023; 23:228. [PMID: 37131145 PMCID: PMC10155446 DOI: 10.1186/s12872-023-03229-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 04/06/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND As a novel marker of insulin resistance, the ratio of triglyceride/high-density lipoprotein cholesterol (TG/HDL-C) has been recently reported to be related to the occurrence of coronary artery diseases. However, no research has been conducted to probe whether the TG/HDL-C ratio is associated with the occurrence of coronary microvascular disease (CMVD). AIM This study investigates the association between the TG/HDL-C ratio and the occurrence of CMVD. METHODS This study included 175 patients diagnosed with CMVD in the Department of Cardiology of our hospital from October 2017 to October 2021 as the study group and 175 patients with no chest pain, no history of cardiovascular disease and drug use, and negative results of exercise treadmill testing as the non-CMVD group. The clinical data of the two groups were compared. In addition, the risk factors of CMVD were analyzed with logistic regression, and the efficacy of independent risk factors in predicting CMVD was analyzed with a receiver operating characteristic (ROC) curve. RESULTS Compared with those in the non-CMVD group, the proportion of females, the incidence of hypertension and type 2 diabetes, the level of platelet count, TG, and C-reactive protein, and the ratio of TG/HDL-C were increased in the CMVD group, accompanied by decreased levels of albumin and HDL-C (P < 0.05). Logistic regression results revealed C-reactive protein (the area under the ROC curve [AUC] value: 0.754; 95% confidence interval [CI]: 0.681-0.827), sex (the AUC value: 0.651; 95%CI: 0.571-0.730), albumin (the AUC value: 0.722; 95%CI: 0.649-0.794), and TG/HDL-C ratio (the AUC value: 0.789; 95%CI: 0.718-0.859) as the independent risk factors of CMVD. CONCLUSION The TG/HDL-C ratio is an independent risk factor for the occurrence of CMVD.
Collapse
Affiliation(s)
- Li Ping Liao
- Cardiology Department, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 800 Huangjiahuayuan Road, Shanghai, 201803, China.
| | - Lei Wu
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, China
| | - Yang Yang
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, China
| |
Collapse
|
44
|
Mathias LS, Herman-de-Sousa C, Cury SS, Nogueira CR, Correia-de-Sá P, de Oliveira M. RNA-seq reveals that anti-obesity irisin and triiodothyronine (T3) hormones differentially affect the purinergic signaling transcriptomics in differentiated human adipocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159276. [PMID: 36642213 DOI: 10.1016/j.bbalip.2022.159276] [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: 07/14/2022] [Revised: 12/05/2022] [Accepted: 12/15/2022] [Indexed: 01/15/2023]
Abstract
The anti-obesity thyroid hormone, triiodothyronine (T3), and irisin, an exercise- and/or cold-induced myokine, stimulate thermogenesis and energy consumption while decreasing lipid accumulation. The involvement of ATP signaling in adipocyte cell function and obesity has attracted increasing attention, but the crosstalk between the purinergic signaling cascade and anti-obesity hormones lacks experimental evidence. In this study, we investigated the effects of T3 and irisin in the transcriptomics of membrane-bound purinoceptors, ectonucleotidase enzymes and nucleoside transporters participating in the purinergic signaling in cultured human adipocytes. The RNA-seq analysis revealed that differentiated adipocytes express high amounts of ADORA1, P2RY11, P2RY12, and P2RX6 gene transcripts, along with abundant levels of transcriptional products encoding to purine metabolizing enzymes (ENPP2, ENPP1, NT5E, ADA and ADK) and transporters (SLC29A1, SCL29A2). The transcriptomics of purinergic signaling markers changed in parallel to the upsurge of "browning" adipocyte markers, like UCP1 and P2RX5, after treatment with T3 and irisin. Upregulation of ADORA1, ADORA2A and P2RX4 gene transcription was obtained with irisin, whereas T3 preferentially upregulated NT5E, SLC29A2 and P2RY11 genes. Irisin was more powerful than T3 towards inhibition of the leptin gene transcription, the SCL29A1 gene encoding for the ENT1 transporter, the E-NPP2 (autotaxin) gene, and genes that encode for two ADP-sensitive P2Y receptors, P2RY1 and P2RY12. These findings indicate that anti-obesity irisin and T3 hormones differentially affect the purinergic signaling transcriptomics, which might point towards new directions for the treatment of obesity and related metabolic disorders that are worth to be pursued in future functional studies.
Collapse
Affiliation(s)
- Lucas Solla Mathias
- Department of Internal Clinic, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Carina Herman-de-Sousa
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal
| | - Sarah Santiloni Cury
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Célia Regina Nogueira
- Department of Internal Clinic, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), ICBAS-UP, Porto, Portugal.
| | - Miriane de Oliveira
- Department of Internal Clinic, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
45
|
Ibáñez L, de Zegher F. Adolescent PCOS: a postpubertal central obesity syndrome. Trends Mol Med 2023; 29:354-363. [PMID: 36964058 DOI: 10.1016/j.molmed.2023.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 03/26/2023]
Abstract
Adolescent polycystic ovary syndrome (PCOS) is a highly prevalent, reversible, endocrine-metabolic mode essentially driven by ectopic fat, which, in turn, often results from a mismatch between early adipogenesis and later lipogenesis, or between prenatal and postnatal weight gain. The key features of adolescent PCOS are menstrual irregularity and androgen excess (hirsutism, acne, and/or high testosterone). Adolescent PCOS is frequently preceded by rapid maturation (early variants of adrenarche/pubarche and puberty/menarche, also accelerated by ectopic fat) and is diagnosed between 2 and 8 years after menarche, thus during late adolescence or early adulthood. Treatment of adolescent PCOS should not only focus on symptoms, but also reduce the amount of ectopic fat, thereby aiming for an overall state of preconception health.
Collapse
Affiliation(s)
- Lourdes Ibáñez
- Endocrinology Department, Research Institute Sant Joan de Déu, University of Barcelona, 08950 Esplugues, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Francis de Zegher
- Leuven Research and Development, University of Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
46
|
Lin F, Liu Y, Rudeski-Rohr T, Dahir N, Calder A, Gilbertson TA. Adiponectin Enhances Fatty Acid Signaling in Human Taste Cells by Increasing Surface Expression of CD36. Int J Mol Sci 2023; 24:ijms24065801. [PMID: 36982874 PMCID: PMC10059208 DOI: 10.3390/ijms24065801] [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: 02/24/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Adiponectin, a key metabolic hormone, is secreted into the circulation by fat cells where it enhances insulin sensitivity and stimulates glucose and fatty acid metabolism. Adiponectin receptors are highly expressed in the taste system; however, their effects and mechanisms of action in the modulation of gustatory function remain unclear. We utilized an immortalized human fungiform taste cell line (HuFF) to investigate the effect of AdipoRon, an adiponectin receptor agonist, on fatty acid-induced calcium responses. We showed that the fat taste receptors (CD36 and GPR120) and taste signaling molecules (Gα-gust, PLCβ2, and TRPM5) were expressed in HuFF cells. Calcium imaging studies showed that linoleic acid induced a dose-dependent calcium response in HuFF cells, and it was significantly reduced by the antagonists of CD36, GPR120, PLCβ2, and TRPM5. AdipoRon administration enhanced HuFF cell responses to fatty acids but not to a mixture of sweet, bitter, and umami tastants. This enhancement was inhibited by an irreversible CD36 antagonist and by an AMPK inhibitor but was not affected by a GPR120 antagonist. AdipoRon increased the phosphorylation of AMPK and the translocation of CD36 to the cell surface, which was eliminated by blocking AMPK. These results indicate that AdipoRon acts to increase cell surface CD36 in HuFF cells to selectively enhance their responses to fatty acids. This, in turn, is consistent with the ability of adiponectin receptor activity to alter taste cues associated with dietary fat intake.
Collapse
Affiliation(s)
- Fangjun Lin
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Yan Liu
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Trina Rudeski-Rohr
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Naima Dahir
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Ashley Calder
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Timothy A Gilbertson
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| |
Collapse
|
47
|
Liu X, Yang Y, Shao H, Liu S, Niu Y, Fu L. Globular adiponectin ameliorates insulin resistance in skeletal muscle by enhancing the LKB1-mediated AMPK activation via SESN2. SPORTS MEDICINE AND HEALTH SCIENCE 2023; 5:34-41. [PMID: 36994173 PMCID: PMC10040333 DOI: 10.1016/j.smhs.2022.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/13/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Adiponectin has been demonstrated to be a mediator of insulin sensitivity; however, the underlined mechanisms remain unclear. SESN2 is a stress-inducible protein that phosphorylates AMPK in different tissues. In this study, we aimed to validate the amelioration of insulin resistance by globular adiponectin (gAd) and to reveal the role of SESN2 in the improvement of glucose metabolism by gAd. We used a high-fat diet-induced wild-type and SESN2-/- C57BL/6J insulin resistance mice model to study the effects of six-week aerobic exercise or gAd administration on insulin resistance. In vitro study, C2C12 myotubes were used to determine the potential mechanism by overexpressing or inhibiting SESN2. Similar to exercise, six-week gAd administration decreased fasting glucose, triglyceride and insulin levels, reduced lipid deposition in skeletal muscle and reversed whole-body insulin resistance in mice fed on a high-fat diet. Moreover, gAd enhanced skeletal muscle glucose uptake by activating insulin signaling. However, these effects were diminished in SESN2-/- mice. We found that gAd administration increased the expression of SESN2 and Liver kinase B1 (LKB1) and increased AMPK-T172 phosphorylation in skeletal muscle of wild-type mice, while in SESN2-/- mice, LKB1 expression was also increased but the pAMPK-T172 was unchanged. At the cellular level, gAd increased cellular SESN2 and pAMPK-T172 expression. Immunoprecipitation experiment suggested that SESN2 promoted the formation of complexes of AMPK and LKB1 and hence phosphorylated AMPK. In conclusion, our results revealed that SESN2 played a critical role in gAd-induced AMPK phosphorylation, activation of insulin signaling and skeletal muscle insulin sensitization in mice with insulin resistance.
Collapse
Affiliation(s)
- Xinmeng Liu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Yang Yang
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Heng Shao
- Department of Anatomy and Histology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Sujuan Liu
- Department of Anatomy and Histology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Yanmei Niu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Li Fu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
48
|
Scheidl TB, Brightwell AL, Easson SH, Thompson JA. Maternal obesity and programming of metabolic syndrome in the offspring: searching for mechanisms in the adipocyte progenitor pool. BMC Med 2023; 21:50. [PMID: 36782211 PMCID: PMC9924890 DOI: 10.1186/s12916-023-02730-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 01/09/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND It is now understood that it is the quality rather than the absolute amount of adipose tissue that confers risk for obesity-associated disease. Adipose-derived stem cells give rise to adipocytes during the developmental establishment of adipose depots. In adult depots, a reservoir of progenitors serves to replace adipocytes that have reached their lifespan and for recruitment to increase lipid buffering capacity under conditions of positive energy balance. MAIN: The adipose tissue expandability hypothesis posits that a failure in de novo differentiation of adipocytes limits lipid storage capacity and leads to spillover of lipids into the circulation, precipitating the onset of obesity-associated disease. Since adipose progenitors are specified to their fate during late fetal life, perturbations in the intrauterine environment may influence the rapid expansion of adipose depots that occurs in childhood or progenitor function in established adult depots. Neonates born to mothers with obesity or diabetes during pregnancy tend to have excessive adiposity at birth and are at increased risk for childhood adiposity and cardiometabolic disease. CONCLUSION In this narrative review, we synthesize current knowledge in the fields of obesity and developmental biology together with literature from the field of the developmental origins of health and disease (DOHaD) to put forth the hypothesis that the intrauterine milieu of pregnancies complicated by maternal metabolic disease disturbs adipogenesis in the fetus, thereby accelerating the trajectory of adipose expansion in early postnatal life and predisposing to impaired adipose plasticity.
Collapse
Affiliation(s)
- Taylor B. Scheidl
- Cumming School of Medicine, Calgary, Canada
- Alberta Children’s Hospital Research Institute, Calgary, Canada
- Libin Cardiovascular Institute, Calgary, Canada
- University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1 Canada
| | - Amy L. Brightwell
- University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1 Canada
| | - Sarah H. Easson
- Cumming School of Medicine, Calgary, Canada
- University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1 Canada
| | - Jennifer A. Thompson
- Cumming School of Medicine, Calgary, Canada
- Alberta Children’s Hospital Research Institute, Calgary, Canada
- Libin Cardiovascular Institute, Calgary, Canada
- University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1 Canada
| |
Collapse
|
49
|
Crosstalk between Adipose Tissue and Hepatic Mitochondria in the Development of the Inflammation and Liver Injury during Ageing in High-Fat Diet Fed Rats. Int J Mol Sci 2023; 24:ijms24032967. [PMID: 36769289 PMCID: PMC9917792 DOI: 10.3390/ijms24032967] [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: 11/29/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Obesity is considered an epidemic disorder, due to an imbalance between energy consumption and metabolizable energy intake. This balance is increasingly disrupted during normal aging processes due to the progressive impairment of mechanisms that normally control energy homeostasis. Obesity is triggered by an excessive lipid depots but reflects systemic inflammation along with large adipocytes secreting proinflammatory adipokines, an increase of the free fatty acids levels in the bloodstream, and ectopic lipid accumulation. Hepatic fat accumulation is the most common cause of chronic liver disease, characterized by mitochondrial dysfunction with a consequent impaired fat metabolism and increased oxidative stress. Therefore, mitochondrial dysfunction is associated to hepatic lipid accumulation and related complications. In this study, we assessed the crosstalk between adipose tissue and liver, analyzing the time-course of changes in hepatic mitochondrial fatty acid oxidation capacity versus fatty acid storage, focusing on the contribution of adipose tissue inflammation to hepatic lipid accumulation, using a rodent model of high fat diet-induced obesity. Our results demonstrate that both high-fat diet-induced obesity and aging induce dysregulation of adipose tissue function and similar metabolic alterations mediated by mitochondrial function impairment and altered inflammatory profile. The high fat diet-induced obesity anticipates and exacerbates liver mitochondrial dysfunction that occurs with aging processes.
Collapse
|
50
|
Could Naringenin Participate as a Regulator of Obesity and Satiety? Molecules 2023; 28:molecules28031450. [PMID: 36771113 PMCID: PMC9921626 DOI: 10.3390/molecules28031450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Obesity is a serious health problem worldwide, since it is associated with multiple metabolic disorders and complications such as cardiovascular disease, type 2 diabetes, fatty liver disease and overall metabolic dysfunction. Dysregulation of the hunger-satiety pathway, which includes alterations of central and peripheral signaling, explains some forms of obesity by favoring hyperphagia and weight gain. The present work comprehensively summarizes the mechanisms by which naringenin (NAR), a predominant flavanone in citrus fruits, could modulate the main pathways associated with the development of obesity and some of its comorbidities, such as oxidative stress (OS), inflammation, insulin resistance (IR) and dyslipidemia, as well as the role of NAR in modulating the secretion of enterohormones of the satiety pathway and its possible antiobesogenic effect. The results of multiple in vitro and in vivo studies have shown that NAR has various potentially modulatory biological effects against obesity by countering IR, inflammation, OS, macrophage infiltration, dyslipidemia, hepatic steatosis, and adipose deposition. Likewise, NAR is capable of modulating peptides or peripheral hormones directly associated with the hunger-satiety pathway, such as ghrelin, cholecystokinin, insulin, adiponectin and leptin. The evidence supports the use of NAR as a promising alternative to prevent overweight and obesity.
Collapse
|