1
|
Liu B, Yao Z, Song L, Sun C, Shen C, Cheng F, Cheng Z, Zhang R, Liu R. Vitexin alleviates lipid metabolism disorders and hepatic injury in obese mice through the PI3K/AKT/mTOR/SREBP-1c pathway. Eur J Med Chem 2025; 287:117379. [PMID: 39947052 DOI: 10.1016/j.ejmech.2025.117379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/24/2025]
Abstract
Obesity is recognized as a metabolic disorder, and its treatment and management pose ongoing challenges worldwide. Hawthorn, a traditional Chinese herb used to alleviate digestive issues and reduce blood lipid levels, has unclear mechanisms of action regarding its active components in the treatment of obesity. This study investigated the anti-obesity effects of vitexin, a major flavonoid compound found in hawthorn, in high-fat diet (HFD)-induced C57BL/6 mice. The results demonstrated that vitexin significantly reduced body weight, liver weight, blood lipid levels, and inflammatory markers in obese mice, while also inhibiting hepatic lipid accumulation. Mechanistic studies revealed that vitexin likely suppresses adipogenesis by modulating the PI3K-AKT signaling pathway, as evidenced by reduced expression of PI3K, phosphorylated AKT, phosphorylated mTOR, and SREBP-1c in the livers of vitexin-treated obese mice. Additionally, vitexin inhibited NFκB expression by regulating IκBα phosphorylation, thereby alleviating obesity-induced liver injury. These findings suggest that vitexin may be the primary active component in hawthorn responsible for reducing blood lipid levels, highlighting its potential in the treatment of obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
- Bo Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ziqing Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lin Song
- Department of Pharmacy, Children' S Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Chen Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Changhong Shen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zefang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ruoqi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Rong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
2
|
Lin YK, Pan YF, Jiang TY, Chen YB, Shang TY, Xu MY, Feng HB, Ma YH, Tan YX, Wang HY, Dong LW. Blocking the SIRPα-CD47 axis promotes macrophage phagocytosis of exosomes derived from visceral adipose tissue and improves inflammation and metabolism in mice. J Biomed Sci 2025; 32:31. [PMID: 40016734 PMCID: PMC11869713 DOI: 10.1186/s12929-025-01124-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/06/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Adipose tissue plays a pivotal role in systemic metabolism and maintaining bodily homeostasis. Exosomes from adipose tissues, known as AT-Exos, are recognized as important messengers in the communication between adipose tissue and other organs. Despite this, the alterations in exosome composition and the functional disparities among depot-specific AT-Exos in obesity remain elusive. METHODS In this work, we utilized lipidomics and microRNA (miRNA) sequencing to elucidate the lipid and miRNA profiles of AT-Exos in a diet-induced obesity model. We identified obesity-related miRNAs in AT-Exos and further explored their mechanisms using gain- and loss-of-function experiments. To evaluate the metabolic effects of AT-Exos on adipocytes, we conducted RNA-sequencing (RNA-seq) and confirmed our findings through Quantitative Real-time PCR (qPCR) and Western bolt analyses. Meanwhile, a mouse model with intraperitoneal injections was utilized to validate the role of exosomes derived from visceral white adipose tissue (vWAT-Exos) in obesity progression in vivo. Finally, we explored potential therapeutic intervention strategies targeting AT-Exos, particularly focusing on modulating the SIRPα-CD47 axis to enhance macrophage phagocytosis using Leptin-deficient (ob/ob) mice and SIRPα knock-out mice. RESULTS Our study revealed that obesity-related metabolism affects the biological processes of AT-Exos, with depot-specific secretion patterns. In obesity, the lipidome profile of AT-Exos was significantly altered, and diet can modify the miRNA content and function within these exosomes, influencing lipid metabolism and inflammatory pathways that contribute to metabolic dysregulation. Specifically, we identified that miR-200a-3p and miR-200b-3p promoted lipid accumulation in 3T3L1 cells partly through the PI3K/AKT/mTOR pathway. RNA-Seq analysis revealed that AT-Exos from different fat depots exerted distinct effects on adipocyte metabolism, with obese vWAT-Exos being notably potent in triggering inflammation and lipid accumulation in diet-induced obesity. Additionally, we found that inhibiting the SIRPα-CD47 axis can mitigate metabolic disorders induced by obese vWAT-Exos or ob/ob mice, partly due to the enhanced clearance of vWAT-Exos. Consistent with this, SIRPα-deficient mice exhibited a reduction in vWAT-Exos and displayed greater resistance to obesity. CONCLUSIONS This study elucidates that diet-induced obesity altered the lipid and miRNA profiles of AT-Exos, which involved in modulating adipocyte inflammation and metabolic balance. The SIRPα-CD47 axis emerges as a potential therapeutic target for obesity and its associated complications.
Collapse
Affiliation(s)
- Yun-Kai Lin
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Yu-Fei Pan
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Tian-Yi Jiang
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Yi-Bin Chen
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Tai-Yu Shang
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Meng-You Xu
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Hui-Bo Feng
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Yun-Han Ma
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Ye-Xiong Tan
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Hong-Yang Wang
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Li-Wei Dong
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China.
| |
Collapse
|
3
|
Liu X, Du P, Xu J, Wang W, Zhang C. Therapeutic Effects of Intermittent Fasting Combined with SLBZS and Prebiotics on STZ-HFD-Induced Type 2 Diabetic Mice. Diabetes Metab Syndr Obes 2024; 17:4013-4030. [PMID: 39492963 PMCID: PMC11531242 DOI: 10.2147/dmso.s474196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Purpose This study aims to assess the therapeutic potential of combining Shen-Ling-Bai-Zhu-San (SLBZS) or prebiotics with intermittent fasting (IF) in type 2 diabetes mellitus (T2DM) mice and to investigate the synergistic effects and underlying mechanisms. Methods Type 2 diabetic mouse models were induced using high-fat diet (HFD) and streptozotocin (STZ), followed by IF treatment. Mice were then grouped for combined therapy with different doses of SLBZS and prebiotics. Fasting blood glucose (FBG) levels, body weight variations, and oral glucose tolerance tests were assessed to elucidate metabolic alterations. The hepatic and renal parameters were evaluated to determine systemic changes in T2DM mice, while the insulin levels were quantified by ELISA to assess glucose homeostasis. Gut microbiota alterations were examined via 16S rRNA sequencing. Alterations of the genes in relevant signaling pathways were analyzed using RT-qPCR. Results IF improved FBG, body weight, insulin levels, and other diabetes indicators. Combined IF with SLBZS or prebiotics yielded similar effects. Furthermore, it ameliorated dyslipidemia and mitigated hepatic and renal parameters in T2DM mice. Pancreatic tissue histopathology showed islet cell restoration post-intervention. IF therapy reduced the abnormally elevated GSK-3β gene expression and increased the abnormally reduced GLUT2 genes. Further analysis indicated that the combination of IF with prebiotics and high doses of SLBZS upregulated the expression of the INSR and IRS1 genes. Gut microbiota analysis revealed restored diversity and structure, with notable changes in specific bacterial families. At the family level, the contents of Akkermansiaceae and Bifidobacteriaceae were restored. Phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt2) analysis suggested metabolic pathway alterations. Conclusion IF improved type 2 diabetic symptoms, with combined SLBZS and prebiotics showing similar effects. IF with high concentration of SLBZS and prebiotics doses upregulated the INSR and IRS1 genes and had superior effects on gut microbiota compared to IF alone.
Collapse
Affiliation(s)
- Xiaoyu Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Pengyun Du
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Jianing Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Wei Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Chenggang Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| |
Collapse
|
4
|
Arniotis-Streat S, Fonte A, Ziauddeen H. Psychotropic drugs, eating behaviour and weight gain. Curr Opin Psychiatry 2024; 37:331-336. [PMID: 38994802 DOI: 10.1097/yco.0000000000000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
PURPOSE OF REVIEW Psychotropic drug related weight gain is a common side-effect of significant concern to both clinicians and patients. Recent studies and treatment guidelines strongly support taking preventive and early treatment approaches to psychotropic drug-related weight gain (PDWG). Arguably the main pathway that PDWG occurs is via changes in eating behaviour leading to increased caloric intake. RECENT FINDINGS Systematic reviews and meta-analyses have provided good data on the nature and prevalence of alterations in eating behaviour with psychotropic treatment including increased hunger, night eating and binge eating. These changes are unsurprisingly more prominent with agents like olanzapine and clozapine that have high propensity to cause weight gain. SUMMARY Altered eating behaviour can serve as an earlier measure of the risk of weight gain and can be examined easily in clinical practice. Detecting these changes can enable earlier action in terms of switching treatments and starting pharmacological and nonpharmacological preventive strategies.
Collapse
Affiliation(s)
| | - Anthony Fonte
- Mental Health service, Fiona Stanley Fremantle Hospitals Group, Murdoch, WA, Australia
| | - Hisham Ziauddeen
- Mental Health service, Fiona Stanley Fremantle Hospitals Group, Murdoch, WA, Australia
- Dept of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
5
|
Borges KS, Little DW, Magalhães TDA, Ribeiro C, Dumontet T, Lapensee C, Basham KJ, Seth A, Azova S, Guagliardo NA, Barrett PQ, Berber M, O'Connell AE, Turcu AF, Lerario AM, Mohan DR, Rainey W, Carlone DL, Hirschhorn JN, Salic A, Breault DT, Hammer GD. Non-canonical Wnt signaling triggered by WNT2B drives adrenal aldosterone production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609423. [PMID: 39229119 PMCID: PMC11370552 DOI: 10.1101/2024.08.23.609423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The steroid hormone aldosterone, produced by the zona glomerulosa (zG) of the adrenal gland, is a master regulator of plasma electrolytes and blood pressure. While aldosterone control by the renin-angiotensin system is well understood, other key regulatory factors have remained elusive. Here, we replicated a prior association between a non-coding variant in WNT2B and an increased risk of primary aldosteronism, a prevalent and debilitating disease caused by excessive aldosterone production. We further show that in both mice and humans, WNT2B is expressed in the mesenchymal capsule surrounding the adrenal cortex, in close proximity to the zG. Global loss of Wnt2b in the mouse results in a dysmorphic and hypocellular zG, with impaired aldosterone production. Similarly, humans harboring WNT2B loss-of-function mutations develop a novel form of Familial Hyperreninemic Hypoaldosteronism, designated here as Type 4. Additionally, we demonstrate that WNT2B signals by activating the non-canonical Wnt/planar cell polarity pathway. Our findings identify WNT2B as a key regulator of zG function and aldosterone production with important clinical implications.
Collapse
Affiliation(s)
- Kleiton S Borges
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Donald W Little
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Claudio Ribeiro
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Typhanie Dumontet
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chris Lapensee
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Aishwarya Seth
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
| | - Svetlana Azova
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Nick A Guagliardo
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0735, USA
| | - Paula Q Barrett
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0735, USA
| | - Mesut Berber
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Amy E O'Connell
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Adina F Turcu
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Antonio Marcondes Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dipika R Mohan
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - William Rainey
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Joel N Hirschhorn
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
6
|
Kühl F, Brand K, Lichtinghagen R, Huber R. GSK3-Driven Modulation of Inflammation and Tissue Integrity in the Animal Model. Int J Mol Sci 2024; 25:8263. [PMID: 39125833 PMCID: PMC11312333 DOI: 10.3390/ijms25158263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Nowadays, GSK3 is accepted as an enzyme strongly involved in the regulation of inflammation by balancing the pro- and anti-inflammatory responses of cells and organisms, thus influencing the initiation, progression, and resolution of inflammatory processes at multiple levels. Disturbances within its broad functional scope, either intrinsically or extrinsically induced, harbor the risk of profound disruptions to the regular course of the immune response, including the formation of severe inflammation-related diseases. Therefore, this review aims at summarizing and contextualizing the current knowledge derived from animal models to further shape our understanding of GSK3α and β and their roles in the inflammatory process and the occurrence of tissue/organ damage. Following a short recapitulation of structure, function, and regulation of GSK3, we will focus on the lessons learned from GSK3α/β knock-out and knock-in/overexpression models, both conventional and conditional, as well as a variety of (predominantly rodent) disease models reflecting defined pathologic conditions with a significant proportion of inflammation and inflammation-related tissue injury. In summary, the literature suggests that GSK3 acts as a crucial switch driving pro-inflammatory and destructive processes and thus contributes significantly to the pathogenesis of inflammation-associated diseases.
Collapse
Affiliation(s)
| | | | | | - René Huber
- Institute of Clinical Chemistry and Laboratory Medicine, Hannover Medical School, 30625 Hannover, Germany; (F.K.); (K.B.); (R.L.)
| |
Collapse
|
7
|
El-Sayed NS, Khalil NA, Saleh SR, Aly RG, Basta M. The Possible Neuroprotective Effect of Caffeic Acid on Cognitive Changes and Anxiety-Like Behavior Occurring in Young Rats Fed on High-Fat Diet and Exposed to Chronic Stress: Role of β-Catenin/GSK-3B Pathway. J Mol Neurosci 2024; 74:61. [PMID: 38954245 DOI: 10.1007/s12031-024-02232-4] [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: 01/09/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024]
Abstract
Lifestyle influences physical and cognitive development during the period of adolescence greatly. The most important of these lifestyle factors are diet and stress. Therefore, the aim of this study was to investigate the impact of high fat diet (HFD) and chronic mild stress on cognitive function and anxiety-like behaviors in young rats and to study the role of caffeic acid as a potential treatment for anxiety and cognitive dysfunction. Forty rats were assigned into 4 groups: control, HFD, HFD + stress, and caffeic acid-treated group. Rats were sacrificed after neurobehavioral testing. We detected memory impairment and anxiety-like behavior in rats which were more exaggerated in stressed rats. Alongside the behavioral changes, there were biochemical and histological changes. HFD and/or stress decreased hippocampal brain-derived neurotrophic factor (BDNF) levels and induced oxidative and inflammatory changes in the hippocampus. In addition, they suppressed Wnt/β-catenin pathway which was associated with activation of glycogen synthase kinase 3β (GSK3β). HFD and stress increased arginase 1 and inducible nitric oxide synthase (iNOS) levels as well. These disturbances were found to be aggravated in stressed rats than HFD group. However, caffeic acid was able to reverse these deteriorations leading to memory improvement and ameliorating anxiety-like behavior. So, the current study highlights an important neuroprotective role for caffeic acid that may guard against induction of cognitive dysfunction and anxiety disorders in adolescents who are exposed to HFD and/or stress.
Collapse
Affiliation(s)
- Norhan S El-Sayed
- Department of Medical Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt.
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Nehal Adel Khalil
- Department of Medical Biochemistry, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Samar R Saleh
- Department of Biochemistry, Faculty of Science, Alexandria University, Baghdad St., Moharam Bek, Alexandria, 21511, Egypt
- Bioscreening and Preclinical Trial Lab, Department of Biochemistry, Faculty of Science, Alexandria University, Baghdad St., Moharam Bek, Alexandria, 21511, Egypt
| | - Rania G Aly
- Department of pathology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Marianne Basta
- Department of Medical Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| |
Collapse
|
8
|
Zhang B, Feng H, Lin H, Li R. Somatostatin-SSTR3-GSK3 modulates human T-cell responses by inhibiting OXPHOS. Front Immunol 2024; 15:1322670. [PMID: 38426092 PMCID: PMC10902055 DOI: 10.3389/fimmu.2024.1322670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Somatostatin (SST) is a peptide hormone primarily synthesized in the digestive and nervous systems. While its impact on the endocrine system is well-established, accumulating evidence suggests a crucial role for SST and its analogues in modulating immune responses. Despite this, the precise mechanism through which SST regulates T cells has remained largely unknown. Methods To elucidate the impact of SST on human T cells, we conducted a series of experiments involving cell culture assays, molecular analyses, and metabolic profiling. Human T cells were treated with SST, and various parameters including proliferation, cytokine production, and metabolic activities were assessed. Additionally, we employed pharmacological inhibitors and genetic manipulations to dissect the signaling pathways mediating SST's effects on T cells. Results We showed that SST diminishes T-cell proliferation by influencing IL-2 production and T-cell mitochondrial respiration, while having no discernible impact on TCR-induced glycolysis. Our findings also identified that the regulatory influence of SST on T-cell responses and metabolism is contingent on its receptor, SSTR3. Moreover, we demonstrated that SST governs T-cell responses and metabolism by acting through the T-cell metabolic checkpoint GSK3. Discussion Our study provides novel insights into the immunoregulatory function of SST in human T cells, highlighting the complex interplay between hormonal signaling and immune regulation. Understanding the molecular mechanisms underlying SST's effects on T cells may offer therapeutic opportunities for manipulating immune responses in various pathological conditions.
Collapse
Affiliation(s)
- Bo Zhang
- Institute of Neuroscience and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Research, Fujian Medical University, Fuzhou, China
- Institute of Immunotherapy and Department of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Huiru Feng
- Institute of Immunotherapy and Department of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Hui Lin
- Institute of Immunotherapy and Department of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Rui Li
- Institute of Immunotherapy and Department of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
9
|
Engin A. Adiponectin Resistance in Obesity: Adiponectin Leptin/Insulin Interaction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:431-462. [PMID: 39287861 DOI: 10.1007/978-3-031-63657-8_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adiponectin (APN) levels in obesity are negatively correlated with chronic subclinical inflammation markers. The hypertrophic adipocytes cause obesity-linked insulin resistance and metabolic syndrome. Furthermore, macrophage polarization is a key determinant regulating adiponectin receptor (AdipoR1/R2) expression and differential adiponectin-mediated macrophage inflammatory responses in obese individuals. In addition to decrease in adiponectin concentrations, the decline in AdipoR1/R2 messenger ribonucleic acid (mRNA) expression leads to a decrement in adiponectin binding to cell membrane, and this turns into attenuation in the adiponectin effects. This is defined as APN resistance, and it is linked with insulin resistance in high-fat diet-fed subjects. The insulin-resistant group has a significantly higher leptin-to-APN ratio. The leptin-to-APN ratio is more than twofold higher in obese individuals. An increase in expression of AdipoRs restores insulin sensitivity and β-oxidation of fatty acids via triggering intracellular signal cascades. The ratio of high molecular weight to total APN is defined as the APN sensitivity index (ASI). This index is correlated to insulin sensitivity. Homeostasis model of assessment (HOMA)-APN and HOMA-estimated insulin resistance (HOMA-IR) are the most suitable methods to estimate the metabolic risk in metabolic syndrome. While morbidly obese patients display a significantly higher plasma leptin and soluble (s)E-selectin concentrations, leptin-to-APN ratio, there is a significant negative correlation between leptin-to-APN ratio and sP-selectin in obese patients. When comparing the metabolic dysregulated obese group with the metabolically healthy obese group, postprandial triglyceride clearance, insulin resistance, and leptin resistance are significantly delayed following the oral fat tolerance test in the first group. A neuropeptide, Spexin (SPX), is positively correlated with the quantitative insulin sensitivity check index (QUICKI) and APN. APN resistance together with insulin resistance forms a vicious cycle. Despite normal or high APN levels, an impaired post-receptor signaling due to adaptor protein-containing pleckstrin homology domain, phosphotyrosine-binding domain, and leucine zipper motif 1 (APPL1)/APPL2 may alter APN efficiency and activity. However, APPL2 blocks adiponectin signaling through AdipoR1 and AdipoR2 because of the competitive inhibition of APPL1. APPL1, the intracellular binding partner of AdipoRs, is also an important mediator of adiponectin-dependent insulin sensitization. The elevated adiponectin levels with adiponectin resistance are compensatory responses in the condition of an unusual discordance between insulin resistance and APN unresponsiveness. Hypothalamic recombinant adeno-associated virus (rAAV)-leptin (Lep) gene therapy reduces serum APN levels, and it is a more efficient strategy for long-term weight maintenance.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
10
|
Umbarkar P, Ruiz Ramirez SY, Toro Cora A, Tousif S, Lal H. GSK-3 at the heart of cardiometabolic diseases: Isoform-specific targeting is critical to therapeutic benefit. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166724. [PMID: 37094727 PMCID: PMC10247467 DOI: 10.1016/j.bbadis.2023.166724] [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: 01/23/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/26/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a family of serine/threonine kinases. The GSK-3 family has 2 isoforms, GSK-3α and GSK-3β. The GSK-3 isoforms have been shown to play overlapping as well as isoform-specific-unique roles in both, organ homeostasis and the pathogenesis of multiple diseases. In the present review, we will particularly focus on expanding the isoform-specific role of GSK-3 in the pathophysiology of cardiometabolic disorders. We will highlight recent data from our lab that demonstrated the critical role of cardiac fibroblast (CF) GSK-3α in promoting injury-induced myofibroblast transformation, adverse fibrotic remodeling, and deterioration of cardiac function. We will also discuss studies that found the exact opposite role of CF-GSK-3β in cardiac fibrosis. We will review emerging studies with inducible cardiomyocyte (CM)-specific as well as global isoform-specific GSK-3 KOs that demonstrated inhibition of both GSK-3 isoforms provides benefits against obesity-associated cardiometabolic pathologies. The underlying molecular interactions and crosstalk among GSK-3 and other signaling pathways will be discussed. We will briefly review the specificity and limitations of the available small molecule inhibitors targeting GSK-3 and their potential applications to treat metabolic disorders. Finally, we will summarize these findings and offer our perspective on envisioning GSK-3 as a therapeutic target for the management of cardiometabolic diseases.
Collapse
Affiliation(s)
- Prachi Umbarkar
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Sulivette Y Ruiz Ramirez
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Angelica Toro Cora
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Sultan Tousif
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Hind Lal
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
11
|
Zhao X, Li K, Chen M, Liu L. Metabolic codependencies in the tumor microenvironment and gastric cancer: Difficulties and opportunities. Biomed Pharmacother 2023; 162:114601. [PMID: 36989719 DOI: 10.1016/j.biopha.2023.114601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Oncogenesis and the development of tumors affect metabolism throughout the body. Metabolic reprogramming (also known as metabolic remodeling) is a feature of malignant tumors that is driven by oncogenic changes in the cancer cells themselves as well as by cytokines in the tumor microenvironment. These include endothelial cells, matrix fibroblasts, immune cells, and malignant tumor cells. The heterogeneity of mutant clones is affected by the actions of other cells in the tumor and by metabolites and cytokines in the microenvironment. Metabolism can also influence immune cell phenotype and function. Metabolic reprogramming of cancer cells is the result of a convergence of both internal and external signals. The basal metabolic state is maintained by internal signaling, while external signaling fine-tunes the metabolic process based on metabolite availability and cellular needs. This paper reviews the metabolic characteristics of gastric cancer, focusing on the intrinsic and extrinsic mechanisms that drive cancer metabolism in the tumor microenvironment, and interactions between tumor cell metabolic changes and microenvironment metabolic changes. This information will be helpful for the individualized metabolic treatment of gastric cancers.
Collapse
|
12
|
Targeting PI3K/AKT signaling pathway in obesity. Biomed Pharmacother 2023; 159:114244. [PMID: 36638594 DOI: 10.1016/j.biopha.2023.114244] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023] Open
Abstract
Obesity is a disorder with an increasing prevalence, which impairs the life quality of patients and intensifies societal health care costs. The development of safe and innovative prevention strategies and therapeutic approaches is thus of great importance. The complex pathophysiology of obesity involves multiple signaling pathways that influence energy metabolism in different tissues. The phosphatidylinositol 3-kinases (PI3K)/protein kinase B (AKT) pathway is critical for the metabolic homeostasis and its function in insulin-sensitive tissues is described in the context of health, obesity and obesity-related complications. The PI3K family participates in the regulation of diverse physiological processes including but not limited to cell growth, survival, differentiation, autophagy, chemotaxis, and metabolism depending on the cellular context. AKT is downstream of PI3K in the insulin signaling pathway, and promotes multiple cellular processes by targeting a plethora of regulatory proteins that control glucose and lipid metabolism. Natural products are essential for prevention and treatment of many human diseases, including obesity. Anti-obesity natural compounds effect multiple pathophysiological mechanisms involved in obesity development. Numerous recent preclinical studies reveal the advances in using plant secondary metabolites to target the PI3K/AKT signaling pathway for obesity management. In this paper the druggability of PI3K as a target for compounds with anti-obesity potential is evaluated. Perspectives on the strategies and limitations for clinical implementation of obesity management using natural compounds modulating the PI3K/AKT pathway are suggested.
Collapse
|
13
|
Liu X, Yu Z, Zhou HH, Feng Y, Bu Y, Zhai D, Zhang G, Ding S, Wang E, Mi Y, Wan Z. Effect of flavonoid intake on circulating levels of adiponectin and leptin: A systematic review and meta-analysis of randomized controlled clinical trials. Phytother Res 2022; 36:4139-4154. [PMID: 36117321 DOI: 10.1002/ptr.7617] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 11/09/2022]
Abstract
This meta-analysis of randomized controlled trials (RCTs) was conducted to explore the effects of flavonoid intake on adiponectin and leptin levels. The PubMed, EMBASE, and Cochrane Library databases were searched on March 1, 2021. Random-effects, subgroup, sensitivity, and meta-regression analyses were conducted on 40 publications. Flavonoid intake significantly increased circulating adiponectin (0.54 μg/ml, 95% CI [0.20, 0.88], p = .002; I2 = 86.4%) and significantly reduced leptin levels (weighted mean difference: -0.79 ng/ml, 95% CI [-1.33, -0.25], p = .004; I2 = 87.7%). Subgroup analysis demonstrated that flavonoid intervention produced a significant elevation in adiponectin levels only in studies that lasted more than 12 weeks, conducted in Asian regions, were parallel-designed, involved obese or overweight participants and participants with type 2 diabetes mellitus (T2DM) or cardiovascular diseases, used tea catechins, and used a dietary supplement intervention. A significantly negative effect on leptin levels was observed in studies conducted in Asian countries, with healthy participants and participants with T2DM, used whole food interventions, and involved participants with lower baseline leptin levels. In conclusion, flavonoid intake significantly increased circulating adiponectin and decreased leptin levels; however, study heterogeneity was very high. Future well-designed trials are required to address heterogeneous study designs and clarify the efficacy of plants in regulating adiponectin and leptin levels.
Collapse
Affiliation(s)
- Xinxin Liu
- Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Zengli Yu
- College of Public Health, Zhengzhou University, Zhengzhou, China.,NHC Key Laboratory of Birth Defects Prevention & Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Huan-Huan Zhou
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yang Feng
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yongjun Bu
- NHC Key Laboratory of Birth Defects Prevention & Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Desheng Zhai
- NHC Key Laboratory of Birth Defects Prevention & Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Guofu Zhang
- NHC Key Laboratory of Birth Defects Prevention & Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Shibin Ding
- NHC Key Laboratory of Birth Defects Prevention & Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Erhui Wang
- NHC Key Laboratory of Birth Defects Prevention & Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Yang Mi
- Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongxiao Wan
- College of Public Health, Zhengzhou University, Zhengzhou, China.,School of Public Health, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
14
|
Signaling pathways in obesity: mechanisms and therapeutic interventions. Signal Transduct Target Ther 2022; 7:298. [PMID: 36031641 PMCID: PMC9420733 DOI: 10.1038/s41392-022-01149-x] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 12/19/2022] Open
Abstract
Obesity is a complex, chronic disease and global public health challenge. Characterized by excessive fat accumulation in the body, obesity sharply increases the risk of several diseases, such as type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease, and is linked to lower life expectancy. Although lifestyle intervention (diet and exercise) has remarkable effects on weight management, achieving long-term success at weight loss is extremely challenging, and the prevalence of obesity continues to rise worldwide. Over the past decades, the pathophysiology of obesity has been extensively investigated, and an increasing number of signal transduction pathways have been implicated in obesity, making it possible to fight obesity in a more effective and precise way. In this review, we summarize recent advances in the pathogenesis of obesity from both experimental and clinical studies, focusing on signaling pathways and their roles in the regulation of food intake, glucose homeostasis, adipogenesis, thermogenesis, and chronic inflammation. We also discuss the current anti-obesity drugs, as well as weight loss compounds in clinical trials, that target these signals. The evolving knowledge of signaling transduction may shed light on the future direction of obesity research, as we move into a new era of precision medicine.
Collapse
|
15
|
Chen M, Yang Z, Gan H, Wang Y, Li C, Gao Y. Investigation into potential mechanisms of metabolic syndrome by integrative analysis of metabolomics and proteomics. PLoS One 2022; 17:e0270593. [PMID: 35789338 PMCID: PMC9255766 DOI: 10.1371/journal.pone.0270593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 06/14/2022] [Indexed: 11/18/2022] Open
Abstract
Metabolic syndrome (MetS) is a complex syndrome cluster of metabolic disorders, which greatly increases the risks of diabetic and cardiovascular diseases. Although it has become a significantly worldwide public health burden, its pathogenesis largely remains unknown. In this study, we first performed an integrated analysis of proteomic and metabonomic data of liver tissues of rats between MetS and control groups to reveal possible mechanisms of MetS. A total of 16 significantly perturbed pathways were identified, of which three pathways were shared by patients with MetS and diabetes identified by analysis of serum samples, including alanine, aspartate and glutamate metabolism, valine, leucine and isoleucine biosynthesis, and glycine, serine and threonine metabolism. Additionally, it was found that 18 differential metabolites were closely related with 36 differential proteins, which were considered as significantly discriminant metabolites and proteins between two groups and were mainly involved in metabolic processes of gamma-aminobutyric acid and acetyl-CoA, biosynthetic processes of cholesterol and amino acids. The results of PPI network analysis and topological parameter calculation of four methods revealed that 16 proteins can serve as hub proteins of MetS. Followed by searching the PubMed database and molecular docking of Cyp7a1 and Got1, we concluded that atorvastatin and resveratrol may be potential drugs for MetS.
Collapse
Affiliation(s)
- Meimei Chen
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Engineering Research Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Zhaoyang Yang
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Engineering Research Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Huijian Gan
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Engineering Research Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yang Wang
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Engineering Research Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Chandong Li
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Engineering Research Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yuxing Gao
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
16
|
Bala A, Roy S, Das D, Marturi V, Mondal C, Patra S, Haldar PK, Samajdar G. Role of Glycogen Synthase Kinase-3 in the Etiology of Type 2 Diabetes Mellitus: A Review. Curr Diabetes Rev 2022; 18:e300721195147. [PMID: 34376135 DOI: 10.2174/1573399817666210730094225] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/15/2021] [Accepted: 04/26/2021] [Indexed: 11/22/2022]
Abstract
The risk of type 2 diabetes mellitus (T2DM) is increasing abundantly due to lifestyle-related obesity and associated cardiovascular problems. Presently, Glycogen synthase kinase-3 (GSK-3) has gained considerable attention from biomedical scientists to treat diabetes. Phosphorylation of GSK-3 permits a number of cellular activities like regulation of cell signaling, cellular metabolism, cell proliferation and cellular transport. Inhibiting GSK-3 activity by pharmacological intervention has become an important strategy for the management of T2DM. This review focuses on the schematic representation of fundamental GSK-3 enzymology and encompasses the GSK-3 inhibitors as a future therapeutic lead target for the management of T2DM that may significantly regulate insulin sensitivity to insulin receptor, glycogen synthesis and glucose metabolism. The various signaling mechanisms of inhibiting the GSK-3 by describing insulin signaling through Insulin Receptor Substrate (IRS-1), Phosphatidylinositol-3 Kinase (PI3K) and Protein Kinase B (PKB/ AKT) pathways that may hopefully facilitate the pharmacologist to design for antidiabetic drug evaluation model in near future have also been highlighted.
Collapse
Affiliation(s)
- Asis Bala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, EPIP Campus, Hajipur, Bihar 844102, India
- Division of Pharmacology and Toxicology, Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Road, Panihati, Sodepur, Kolkata-700114; India
| | - Susmita Roy
- Division of Pharmacology and Toxicology, Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Road, Panihati, Sodepur, Kolkata-700114; India
| | - Debanjana Das
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, EPIP Campus, Hajipur, Bihar 844102, India
| | - Venkatesh Marturi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, EPIP Campus, Hajipur, Bihar 844102, India
| | - Chaitali Mondal
- TCG Life Sciences (Chembiotek) Pvt. Ltd., Sector V, Salt Lake Electronics Complex, Kolkata, West Bengal 700091, India
| | - Susmita Patra
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Pallab Kanti Haldar
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Gourav Samajdar
- Division of Pharmacology and Toxicology, Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Road, Panihati, Sodepur, Kolkata-700114; India
| |
Collapse
|
17
|
Wang L, Li J, Di LJ. Glycogen synthesis and beyond, a comprehensive review of GSK3 as a key regulator of metabolic pathways and a therapeutic target for treating metabolic diseases. Med Res Rev 2021; 42:946-982. [PMID: 34729791 PMCID: PMC9298385 DOI: 10.1002/med.21867] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/01/2021] [Accepted: 10/24/2021] [Indexed: 12/19/2022]
Abstract
Glycogen synthase kinase‐3 (GSK3) is a highly evolutionarily conserved serine/threonine protein kinase first identified as an enzyme that regulates glycogen synthase (GS) in response to insulin stimulation, which involves GSK3 regulation of glucose metabolism and energy homeostasis. Both isoforms of GSK3, GSK3α, and GSK3β, have been implicated in many biological and pathophysiological processes. The various functions of GSK3 are indicated by its widespread distribution in multiple cell types and tissues. The studies of GSK3 activity using animal models and the observed effects of GSK3‐specific inhibitors provide more insights into the roles of GSK3 in regulating energy metabolism and homeostasis. The cross‐talk between GSK3 and some important energy regulators and sensors and the regulation of GSK3 in mitochondrial activity and component function further highlight the molecular mechanisms in which GSK3 is involved to regulate the metabolic activity, beyond its classical regulatory effect on GS. In this review, we summarize the specific roles of GSK3 in energy metabolism regulation in tissues that are tightly associated with energy metabolism and the functions of GSK3 in the development of metabolic disorders. We also address the impacts of GSK3 on the regulation of mitochondrial function, activity and associated metabolic regulation. The application of GSK3 inhibitors in clinical tests will be highlighted too. Interactions between GSK3 and important energy regulators and GSK3‐mediated responses to different stresses that are related to metabolism are described to provide a brief overview of previously less‐appreciated biological functions of GSK3 in energy metabolism and associated diseases through its regulation of GS and other functions.
Collapse
Affiliation(s)
- Li Wang
- Proteomics, Metabolomics, and Drug Development Core, Faculty of Health Sciences, University of Macau, Macau, China.,Department of Biomedical Sciences, Faculty of Health Sciences, Macau, China.,Cancer Center of the Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, University of Macau, Macau, China.,Ministry of Education, Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Jiajia Li
- Department of Biomedical Sciences, Faculty of Health Sciences, Macau, China.,Cancer Center of the Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, University of Macau, Macau, China.,Ministry of Education, Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Li-Jun Di
- Department of Biomedical Sciences, Faculty of Health Sciences, Macau, China.,Cancer Center of the Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, University of Macau, Macau, China.,Ministry of Education, Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| |
Collapse
|
18
|
Guo C, Gao C, Lv X, Zhao D, Greenaway FT, Hao L, Tian Y, Liu S, Sun M. CRKL promotes hepatocarcinoma through enhancing glucose metabolism of cancer cells via activating PI3K/Akt. J Cell Mol Med 2021; 25:2714-2724. [PMID: 33523562 PMCID: PMC7933966 DOI: 10.1111/jcmm.16303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/05/2020] [Accepted: 01/04/2021] [Indexed: 12/17/2022] Open
Abstract
Abnormal glucose metabolism may contribute to cancer progression. As a member of the CRK (v-crk sarcoma virus CT10 oncogene homologue) adapter protein family, CRKL (CRK-like) associated with the development and progression of various tumours. However, the exact role and underlying mechanism of CRKL on energy metabolism remain unknown. In this study, we investigated the effect of CRKL on glucose metabolism of hepatocarcinoma cells. CRKL and PI3K were found to be overexpressed in both hepatocarcinoma cells and tissues; meanwhile, CRKL up-regulation was positively correlated with PI3K up-regulation. Functional investigations revealed that CRKL overexpression promoted glucose uptake, lactate production and glycogen synthesis of hepatocarcinoma cells by up-regulating glucose transporters 1 (GLUT1), hexokinase II (HKII) expression and down-regulating glycogen synthase kinase 3β (GSK3β) expression. Mechanistically, CRKL promoted glucose metabolism of hepatocarcinoma cells via enhancing the CRKL-PI3K/Akt-GLUT1/HKII-glucose uptake, CRKL-PI3K/Akt-HKII-glucose-lactate production and CRKL-PI3K/Akt-Gsk3β-glycogen synthesis. We demonstrate CRKL facilitates HCC malignancy via enhancing glucose uptake, lactate production and glycogen synthesis through PI3K/Akt pathway. It provides interesting fundamental clues to CRKL-related carcinogenesis through glucose metabolism and offers novel therapeutic strategies for hepatocarcinoma.
Collapse
Affiliation(s)
- Chunmei Guo
- Department of BiotechnologyCollege of Basic Medical SciencesDalian Medical UniversityDalianChina
| | - Chao Gao
- Department of BiotechnologyCollege of Basic Medical SciencesDalian Medical UniversityDalianChina
- Present address:
College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing)DaqingChina
| | - Xinxin Lv
- Department of BiotechnologyCollege of Basic Medical SciencesDalian Medical UniversityDalianChina
| | - Dongting Zhao
- Department of BiotechnologyCollege of Basic Medical SciencesDalian Medical UniversityDalianChina
| | | | - Lihong Hao
- Department of Histology and EmbryologyCollege of Basic Medical SciencesDalian Medical UniversityDalianChina
| | - Yuxiang Tian
- Department of BiochemistryCollege of Basic Medical SciencesDalian Medical UniversityDalianChina
| | - Shuqing Liu
- Department of BiochemistryCollege of Basic Medical SciencesDalian Medical UniversityDalianChina
| | - Ming‐Zhong Sun
- Department of BiotechnologyCollege of Basic Medical SciencesDalian Medical UniversityDalianChina
| |
Collapse
|
19
|
Polito R, Di Meo I, Barbieri M, Daniele A, Paolisso G, Rizzo MR. Adiponectin Role in Neurodegenerative Diseases: Focus on Nutrition Review. Int J Mol Sci 2020; 21:ijms21239255. [PMID: 33291597 PMCID: PMC7729837 DOI: 10.3390/ijms21239255] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Adiponectin is an adipokine produced by adipose tissue. It has numerous beneficial effects. In particular, it improves metabolic effects and glucose homeostasis, lipid profile, and is involved in the regulation of cytokine profile and immune cell production, having anti-inflammatory and immune-regulatory effects. Adiponectin’s role is already known in immune diseases and also in neurodegenerative diseases. Neurodegenerative diseases, such as Alzheimer’s disease and Parkinson’s disease, are a set of diseases of the central nervous system, characterized by a chronic and selective process of neuron cell death, which occurs mainly in relation to oxidative stress and neuroinflammation. Lifestyle is able to influence the development of these diseases. In particular, unhealthy nutrition on gut microbiota, influences its composition and predisposition to develop many diseases such as neurodegenerative diseases, given the importance of the “gut-brain” axis. There is a strong interplay between Adiponectin, gut microbiota, and brain-gut axis. For these reasons, a healthy diet composed of healthy nutrients such as probiotics, prebiotics, polyphenols, can prevent many metabolic and inflammatory diseases such as neurodegenerative diseases and obesity. The special Adiponectin role should be taken into account also, in order to be able to use this component as a therapeutic molecule.
Collapse
Affiliation(s)
- Rita Polito
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza Miraglia 2, 80138 Naples, Italy; (R.P.); (I.D.M.); (M.B.); (G.P.)
- CEINGE-Advanced Biotechnologies Scarl, Via G. Salvatore 486, 80145 Naples, Italy
| | - Irene Di Meo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza Miraglia 2, 80138 Naples, Italy; (R.P.); (I.D.M.); (M.B.); (G.P.)
| | - Michelangela Barbieri
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza Miraglia 2, 80138 Naples, Italy; (R.P.); (I.D.M.); (M.B.); (G.P.)
| | - Aurora Daniele
- Department of Environmental Biological Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Via G. Vivaldi 42, 81100 Caserta, Italy;
- CEINGE-Advanced Biotechnologies Scarl, Via G. Salvatore 486, 80145 Naples, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza Miraglia 2, 80138 Naples, Italy; (R.P.); (I.D.M.); (M.B.); (G.P.)
| | - Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza Miraglia 2, 80138 Naples, Italy; (R.P.); (I.D.M.); (M.B.); (G.P.)
- Correspondence: ; Tel.: +39-081-566-5135; Fax: +39-081-566-5303
| |
Collapse
|
20
|
Tang X, Fang M, Cheng R, Zhang Z, Wang Y, Shen C, Han Y, Lu Q, Du Y, Liu Y, Sun Z, Zhu L, Mwangi J, Xue M, Long C, Lai R. Iron-Deficiency and Estrogen Are Associated With Ischemic Stroke by Up-Regulating Transferrin to Induce Hypercoagulability. Circ Res 2020; 127:651-663. [PMID: 32450779 DOI: 10.1161/circresaha.119.316453] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 05/24/2020] [Indexed: 01/06/2023]
Abstract
RATIONALE Epidemiological studies have identified an associate between iron deficiency (ID) and the use of oral contraceptives (CC) and ischemic stroke (IS). To date, however, the underlying mechanism remains poorly understood. Both ID and CC have been demonstrated to upregulate the level and iron-binding ability of Tf (transferrin), with our recent study showing that this upregulation can induce hypercoagulability by potentiating FXIIa/thrombin and blocking antithrombin-coagulation proteases interactions. OBJECTIVE To investigate whether Tf mediates IS associated with ID or CC and the underlying mechanisms. METHODS AND RESULTS Tf levels were assayed in the plasma of IS patients with a history of ID anemia, ID anemia patients, venous thromboembolism patients using CC, and ID mice, and in the cerebrospinal fluid of some IS patients. Effects of ID and estrogen administration on Tf expression and coagulability and the underlying mechanisms were studied in vivo and in vitro. High levels of Tf and Tf-thrombin/FXIIa complexes were found in patients and ID mice. Both ID and estrogen upregulated Tf through hypoxia and estrogen response elements located in the Tf gene enhancer and promoter regions, respectively. In addition, ID, administration of exogenous Tf or estrogen, and Tf overexpression promoted platelet-based thrombin generation and hypercoagulability and thus aggravated IS. In contrast, anti-Tf antibodies, Tf knockdown, and peptide inhibitors of Tf-thrombin/FXIIa interaction exerted anti-IS effects in vivo. CONCLUSIONS Our findings revealed that certain factors (ie, ID and CC) upregulating Tf are risk factors of thromboembolic diseases decipher a previously unrecognized mechanistic association among ID, CC, and IS and provide a novel strategy for the development of anti-IS medicine by interfering with Tf-thrombin/FXIIa interactions.
Collapse
Affiliation(s)
- Xiaopeng Tang
- From the Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China (X.T., M.F., R.C., Z.Z., C.S., Y.H., Q.L., J.M., M.X., C.L., R.L.)
| | - Mingqian Fang
- From the Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China (X.T., M.F., R.C., Z.Z., C.S., Y.H., Q.L., J.M., M.X., C.L., R.L.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Yunnan, China (M.F., R.C., J.M.)
| | - Ruomei Cheng
- From the Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China (X.T., M.F., R.C., Z.Z., C.S., Y.H., Q.L., J.M., M.X., C.L., R.L.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Yunnan, China (M.F., R.C., J.M.)
| | - Zhiye Zhang
- From the Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China (X.T., M.F., R.C., Z.Z., C.S., Y.H., Q.L., J.M., M.X., C.L., R.L.)
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Kunming, Yunnan, China (Z.Z., Q.L.)
| | - Yuming Wang
- Department of Clinical Laboratory, the Second Affiliated Hospital of Kunming Medical University, Yunnan, China (Y.W.)
| | - Chuanbin Shen
- From the Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China (X.T., M.F., R.C., Z.Z., C.S., Y.H., Q.L., J.M., M.X., C.L., R.L.)
| | - Yajun Han
- From the Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China (X.T., M.F., R.C., Z.Z., C.S., Y.H., Q.L., J.M., M.X., C.L., R.L.)
| | - Qiumin Lu
- From the Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China (X.T., M.F., R.C., Z.Z., C.S., Y.H., Q.L., J.M., M.X., C.L., R.L.)
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Kunming, Yunnan, China (Z.Z., Q.L.)
| | - Yingrong Du
- Department of Cardiology (Y.D.), the Third People's Hospital of Kunming, Yunnan, China
| | - Yingying Liu
- Department of Clinical Laboratory (Y.L.), the Third People's Hospital of Kunming, Yunnan, China
| | - Zhaohui Sun
- Department of Clinical Laboratory, Guangzhou General Hospital of Guangzhou Military Command, Guangdong, China (Z.S.)
| | - Liping Zhu
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, Yunnan, China (L.Z.)
| | - James Mwangi
- From the Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China (X.T., M.F., R.C., Z.Z., C.S., Y.H., Q.L., J.M., M.X., C.L., R.L.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Yunnan, China (M.F., R.C., J.M.)
| | - Min Xue
- From the Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China (X.T., M.F., R.C., Z.Z., C.S., Y.H., Q.L., J.M., M.X., C.L., R.L.)
| | - Chengbo Long
- From the Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China (X.T., M.F., R.C., Z.Z., C.S., Y.H., Q.L., J.M., M.X., C.L., R.L.)
| | - Ren Lai
- From the Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China (X.T., M.F., R.C., Z.Z., C.S., Y.H., Q.L., J.M., M.X., C.L., R.L.)
- Institute for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai, China (R.L.)
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases (R.L.), Kunming Institute of Zoology, Chinese Academy of Sciences, Yunnan, China
- Sino-African Joint Research Center (R.L.), Kunming Institute of Zoology, Chinese Academy of Sciences, Yunnan, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China (R.L.)
| |
Collapse
|
21
|
Identifying potential functional lncRNAs in metabolic syndrome by constructing a lncRNA-miRNA-mRNA network. J Hum Genet 2020; 65:927-938. [PMID: 32690864 DOI: 10.1038/s10038-020-0753-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/18/2020] [Accepted: 03/25/2020] [Indexed: 11/09/2022]
Abstract
The metabolic syndrome (MS) is a cluster of interrelated risk factors including diabetes mellitus, abdominal obesity, high cholesterol, and hypertension, which can significantly increase mortality and disability. Accumulating evidence suggest that long non-coding RNAs (lncRNAs) are involved in the pathogenesis of human metabolic diseases. However, little is known about the regulatory role of lncRNAs in MS. In this work, we proposed a method for identifying potential MS-associated lncRNAs by constructing an lncRNA-miRNA-mRNA network (LMMN). Firstly, we constructed LMMN by integrating MS-associated genes, miRNA-mRNA interactions, miRNA-lncRNA interactions and mRNA/miRNA expression profiles in patients with MS. Then, we predicted potential MS-associated lncRNAs based on the topological properties of LMMN. As a result, we identified XIST as the most important lncRNA in LMMN. Furthermore, we focused on XIST/miR-214-3p and mir-181a-5p/PTEN axis and validated their expression in MS using real-time quantitative polymerase chain reaction (RT-qPCR). The RT-qPCR results showed that the expression of XIST and PTEN was significantly decreased (P < 0.05) while the expression of miR-214-3p was significantly increased (P < 0.05) in peripheral blood mononuclear cells (PBMCs) of patients with MS, compared with healthy controls. In addition, correlation analysis showed that XIST was negatively correlated with serum C peptide and PTEN was positively correlated with BMI of MS patients. Our findings provided new evidence for further exploring the regulatory role of XIST and other lncRNAs in MS.
Collapse
|
22
|
GSK3: A Kinase Balancing Promotion and Resolution of Inflammation. Cells 2020; 9:cells9040820. [PMID: 32231133 PMCID: PMC7226814 DOI: 10.3390/cells9040820] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
GSK3 has been implicated for years in the regulation of inflammation and addressed in a plethora of scientific reports using a variety of experimental (disease) models and approaches. However, the specific role of GSK3 in the inflammatory process is still not fully understood and controversially discussed. Following a detailed overview of structure, function, and various regulatory levels, this review focusses on the immunoregulatory functions of GSK3, including the current knowledge obtained from animal models. Its impact on pro-inflammatory cytokine/chemokine profiles, bacterial/viral infections, and the modulation of associated pro-inflammatory transcriptional and signaling pathways is discussed. Moreover, GSK3 contributes to the resolution of inflammation on multiple levels, e.g., via the regulation of pro-resolving mediators, the clearance of apoptotic immune cells, and tissue repair processes. The influence of GSK3 on the development of different forms of stimulation tolerance is also addressed. Collectively, the role of GSK3 as a kinase balancing the initiation/perpetuation and the amelioration/resolution of inflammation is highlighted.
Collapse
|
23
|
Adiponectin and Cognitive Decline. Int J Mol Sci 2020; 21:ijms21062010. [PMID: 32188008 PMCID: PMC7139651 DOI: 10.3390/ijms21062010] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/10/2020] [Accepted: 03/13/2020] [Indexed: 12/11/2022] Open
Abstract
Adiponectin (ADPN) is a plasma protein secreted by adipose tissue showing pleiotropic effects with anti-diabetic, anti-atherogenic, and anti-inflammatory properties. Initially, it was thought that the main role was only the metabolism control. Later, ADPN receptors were also found in the central nervous system (CNS). In fact, the receptors AdipoR1 and AdipoR2 are expressed in various areas of the brain, including the hypothalamus, hippocampus, and cortex. While AdipoR1 regulates insulin sensitivity through the activation of the AMP-activated protein kinase (AMPK) pathway, AdipoR2 stimulates the neural plasticity through the activation of the peroxisome proliferator-activated receptor alpha (PPARα) pathway that inhibits inflammation and oxidative stress. Overall, based on its central and peripheral actions, ADPN appears to have neuroprotective effects by reducing inflammatory markers, such as C-reactive protein (PCR), interleukin 6 (IL6), and Tumor Necrosis Factor a (TNFa). Conversely, high levels of inflammatory cascade factors appear to inhibit the production of ADPN, suggesting bidirectional modulation. In addition, ADPN appears to have insulin-sensitizing action. It is known that a reduction in insulin signaling is associated with cognitive impairment. Based on this, it is of great interest to investigate the mechanism of restoration of the insulin signal in the brain as an action of ADPN, because it is useful for testing a possible pharmacological treatment for the improvement of cognitive decline. Anyway, if ADPN regulates neuronal functioning and cognitive performances by the glycemic metabolic system remains poorly explored. Moreover, although the mechanism is still unclear, women compared to men have a doubled risk of developing cognitive decline. Several studies have also supported that during the menopausal transition, the estrogen reduction can adversely affect the brain, in particular, verbal memory and verbal fluency. During the postmenopausal period, in obese and insulin-resistant individuals, ADPN serum levels are significantly reduced. Our recent study has evaluated the relationship between plasma ADPN levels and cognitive performances in menopausal women. Thus, the aim of this review is to summarize both the mechanisms and the effects of ADPN in the central nervous system and the relationship between plasma ADPN levels and cognitive performances, also in menopausal women.
Collapse
|
24
|
Recovery of muscle mass and muscle oxidative phenotype following disuse does not require GSK-3 inactivation. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165740. [PMID: 32087280 DOI: 10.1016/j.bbadis.2020.165740] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/24/2020] [Accepted: 02/18/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Physical inactivity contributes to muscle wasting and reductions in mitochondrial oxidative phenotype (OXPHEN), reducing physical performance and quality of life during aging and in chronic disease. Previously, it was shown that inactivation of glycogen synthase kinase (GSK)-3β stimulates muscle protein accretion, myogenesis, and mitochondrial biogenesis. Additionally, GSK-3β is inactivated during recovery of disuse-induced muscle atrophy. AIM Therefore, we hypothesize that GSK-3 inhibition is required for reloading-induced recovery of skeletal muscle mass and OXPHEN. METHODS Wild-type (WT) and whole-body constitutively active (C.A.) Ser21/9 GSK-3α/β knock-in mice were subjected to a 14-day hind-limb suspension/14-day reloading protocol. Soleus muscle mass, fiber cross-sectional area (CSA), OXPHEN (abundance of sub-units of oxidative phosphorylation (OXPHOS) complexes and fiber-type composition), as well as expression levels of their main regulators (respectively protein synthesis/degradation, myogenesis and peroxisome proliferator-activated receptor-γ co-activator-1α (PGC-1α) signaling) were monitored. RESULTS Subtle but consistent differences suggesting suppression of protein turnover signaling and decreased expression of several OXPHOS sub-units and PGC-1α signaling constituents were observed at baseline in C.A. GSK-3 versus WT mice. Although soleus mass recovery during reloading occurred more rapidly in C.A. GSK-3 mice, this was not accompanied by a parallel increased CSA. The OXPHEN response to reloading was not distinct between C.A. GSK-3 and WT mice. No consistent or significant differences in reloading-induced changes in the regulatory steps of protein turnover, myogenesis or muscle OXPHEN were observed in C.A. GSK-3 compared to WT muscle. CONCLUSION This study indicates that GSK-3 inactivation is dispensable for reloading-induced recovery of muscle mass and OXPHEN.
Collapse
|
25
|
Bopp SK, Heilbronner U, Schlattmann P, Mühleisen TW, Bschor T, Richter C, Steinacher B, Stamm TJ, Merkl A, Herms S, Köhler S, Sterzer P, Hellweg R, Heinz A, Cichon S, Lang UE, Schulze TG, Adli M, Ricken R. Leptin gene polymorphisms are associated with weight gain during lithium augmentation in patients with major depression. Eur Neuropsychopharmacol 2019; 29:211-221. [PMID: 30554862 DOI: 10.1016/j.euroneuro.2018.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/29/2018] [Accepted: 12/01/2018] [Indexed: 12/28/2022]
Abstract
Weight gain is a common adverse effect of lithium augmentation. Previous studies indicate an impact of genetic variants at the leptin gene on weight gain as a consequence of psychopharmacological treatment. The primary aim of our study was to identify variants at the leptin locus that might predict lithium-induced weight gain. The secondary aim was to investigate if these variants modulate leptin levels. In 180 patients with acute major depressive disorder, body mass index was measured before and after 4 weeks of lithium augmentation, in a subsample also after 4 and/or 7 months. In a subsample of 89 patients, leptin serum concentrations were measured before and during lithium augmentation. We used linear mixed model analyzes to investigate the effects of 2 polymorphisms at the leptin locus (rs4731426 and rs7799039, employing the respective proxy SNPs rs2278815 and rs10487506) on changes in body mass index and leptin levels. For both polymorphisms, which are in high linkage disequilibrium, body mass index was significantly lower in homozygous A-allele carriers than in carriers of other genotypes at baseline. Over the follow-up period, body mass index increased less in homozygous A-allele carriers of rs4731426 than in carriers of other genotypes. This was not the case for rs7799039. Neither polymorphism modulated leptin protein expression. Our study strongly supports the hypothesis that genetic variability at the leptin locus is involved in lithium augmentation-associated weight gain in major depressive disorder. Furthermore, Genotype-Tissue Expression data provide strong evidence that rs4731426 influences the expression of leptin messenger ribonucleic acid in fibroblasts.
Collapse
Affiliation(s)
- Sandra K Bopp
- Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Urs Heilbronner
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany
| | - Peter Schlattmann
- Department of Statistics, Informatics and Documentation, Friedrich-Schiller-University Jena, Jena, Germany
| | - Thomas W Mühleisen
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany; Human Genomics Research Group and Division of Medical Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Tom Bschor
- Department of Psychiatry, Schlosspark Hospital Berlin, Berlin, Germany; Department of Psychiatry and Psychotherapy, Technical University of Dresden Medical School, Dresden, Germany
| | - Christoph Richter
- Department of Psychiatry and Psychotherapy, Vivantes Hospital, Kaulsdorf, Berlin, Germany; Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Bruno Steinacher
- Department of Psychiatry and Psychotherapy, Vivantes Hospital Wenckebach, Berlin, Germany
| | - Thomas J Stamm
- Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany; Medical School Brandenburg Theodor Fontane, Neuruppin, Germany
| | - Angela Merkl
- Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany; Department of Psychiatry and Psychotherapy, Fliedner Hospital Berlin, Berlin, Germany
| | - Stefan Herms
- Human Genomics Research Group and Division of Medical Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland; Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Stephan Köhler
- Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Philipp Sterzer
- Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Rainer Hellweg
- Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Sven Cichon
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany; Human Genomics Research Group and Division of Medical Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland; Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Undine E Lang
- Department of Psychiatry and Psychotherapy, University Psychiatric Clinics (UPK),University of Basel, Switzerland
| | - Thomas G Schulze
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany; Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Mazda Adli
- Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany; Department of Psychiatry and Psychotherapy, Fliedner Hospital Berlin, Berlin, Germany
| | - Roland Ricken
- Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
26
|
An expanding GSK3 network: implications for aging research. GeroScience 2019; 41:369-382. [PMID: 31313216 DOI: 10.1007/s11357-019-00085-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/02/2019] [Indexed: 10/26/2022] Open
Abstract
The last few decades of longevity research have been very exciting. We now know that longevity and healthspan can be manipulated across species, from unicellular eukaryotes to nonhuman primates, and that while aging itself is inevitable, how we age is malleable. Numerous dietary, genetic, and pharmacological studies now point to links between metabolism and growth regulation as a central aspect in determining longevity and, perhaps more importantly, health with advancing age. Here, we focus on a relatively new player in aging studies GSK3, glycogen synthase kinase, a key factor in growth and metabolism whose name fails to convey the extensive breadth of its role in cellular adaptation. First, we provide a brief overview of GSK3, touching on those aspects that are likely relevant to aging. Then, we outline the role of GSK3 in cellular functions including growth signaling, cell fate, and metabolism. Next, we describe evidence demonstrating a direct role for GSK3 in a range of age-related diseases, despite the fact that they differ considerably in their etiology and pathology. Finally, we discuss the role that GSK3 may play in normative aging and how GSK3 might be a suitable target to oppose age-related disease vulnerability.
Collapse
|
27
|
Abstract
The organs require oxygen and other types of nutrients (amino acids, sugars, and lipids) to function, the heart consuming large amounts of fatty acids for oxidation and adenosine triphosphate (ATP) generation.
Collapse
|
28
|
Tsakiri EN, Gumeni S, Iliaki KK, Benaki D, Vougas K, Sykiotis GP, Gorgoulis VG, Mikros E, Scorrano L, Trougakos IP. Hyperactivation of Nrf2 increases stress tolerance at the cost of aging acceleration due to metabolic deregulation. Aging Cell 2019; 18:e12845. [PMID: 30537423 PMCID: PMC6351879 DOI: 10.1111/acel.12845] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/20/2018] [Accepted: 08/26/2018] [Indexed: 12/22/2022] Open
Abstract
Metazoans viability depends on their ability to regulate metabolic processes and also to respond to harmful challenges by mounting anti‐stress responses; these adaptations were fundamental forces during evolution. Central to anti‐stress responses are a number of short‐lived transcription factors that by functioning as stress sensors mobilize genomic responses aiming to eliminate stressors. We show here that increased expression of nuclear factor erythroid 2‐related factor (Nrf2) in Drosophila activated cytoprotective modules and enhanced stress tolerance. However, while mild Nrf2 activation extended lifespan, high Nrf2 expression levels resulted in developmental lethality or, after inducible activation in adult flies, in altered mitochondrial bioenergetics, the appearance of Diabetes Type 1 hallmarks and aging acceleration. Genetic or dietary suppression of Insulin/IGF‐like signaling (IIS) titrated Nrf2 activity to lower levels, largely normalized metabolic pathways signaling, and extended flies’ lifespan. Thus, prolonged stress signaling by otherwise cytoprotective short‐lived stress sensors perturbs IIS resulting in re‐allocation of resources from growth and longevity to somatic preservation and stress tolerance. These findings provide a reasonable explanation of why most (if not all) cytoprotective stress sensors are short‐lived proteins, and it also explains the build‐in negative feedback loops (shown here for Nrf2); the low basal levels of these proteins, and why their suppressors were favored by evolution.
Collapse
Affiliation(s)
- Eleni N. Tsakiri
- Department of Cell Biology and Biophysics Faculty of Biology National & Kapodistrian University of Athens Athens Greece
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics Faculty of Biology National & Kapodistrian University of Athens Athens Greece
| | - Kalliopi K. Iliaki
- Department of Cell Biology and Biophysics Faculty of Biology National & Kapodistrian University of Athens Athens Greece
| | - Dimitra Benaki
- Department of Pharmaceutical Chemistry Faculty of Pharmacy National & Kapodistrian University of Athens Athens Greece
| | | | - Gerasimos P. Sykiotis
- Service of Endocrinology, Diabetology and Metabolism Lausanne University Hospital Lausanne Switzerland
| | - Vassilis G. Gorgoulis
- Biomedical Research Foundation Academy of Athens Athens Greece
- Department of Histology and Embryology School of Medicine National & Kapodistrian University of Athens Athens Greece
- Faculty of Biology, Medicine and Health University of Manchester Manchester UK
| | - Emmanuel Mikros
- Department of Pharmaceutical Chemistry Faculty of Pharmacy National & Kapodistrian University of Athens Athens Greece
| | - Luca Scorrano
- Department of Biology, Venetian Institute of Molecular Medicine, Dulbecco‐Telethon Institute University of Padua Padova Italy
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics Faculty of Biology National & Kapodistrian University of Athens Athens Greece
| |
Collapse
|
29
|
Yan H, Guo H, Cheng D, Kou R, Zhang C, Si J. Tributyltin reduces the levels of serum adiponectin and activity of AKT and induces metabolic syndrome in male mice. ENVIRONMENTAL TOXICOLOGY 2018; 33:752-758. [PMID: 29675927 DOI: 10.1002/tox.22562] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 03/28/2018] [Accepted: 04/01/2018] [Indexed: 06/08/2023]
Abstract
Tributyltin (TBT), a proven environmental obesogen, functions as a nanomolar agonist of the peroxisome proliferator activated receptor-γ (PPARγ). However, the adverse effects of TBT on metabolism are incompletely understood. In this study, male ICR mice were administered TBT (5 and 50 μg·kg-1 ) by an intraperitoneal injection once every 3 days for 30 days from 28 days of age and bred for another 30 days after the last administration of TBT. We analyzed the effects of these exposures on the fat depot weights, serum lipid profile, serum leptin and adiponectin, hepatic lipid accumulation, and activity of AKT in the liver and skeletal muscle isolated from mice 8 mins after receiving an insulin injection. Pubertal exposure to TBTCl resulted in a higher body weight, increased epididymal and liver fat accumulation, hyperlipidemia, an elevated low-density lipoprotein/high-density lipoprotein ratio, serum adiponectin deficiency, worse glucose tolerance, and lower insulin-dependent AKT phosphorylation in the liver and muscle in mice. These results showed that TBT exposure induced peripheral insulin resistance and metabolic syndrome in mice.
Collapse
Affiliation(s)
- Haotian Yan
- Department of Environmental Health, School of Public Health, Shandong University, Jinan, China
| | - Hao Guo
- Department of Environmental Health, School of Public Health, Shandong University, Jinan, China
| | - Dong Cheng
- Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, Shandong, 250014, China
| | - Ruirui Kou
- Department of Environmental Health, School of Public Health, Shandong University, Jinan, China
| | - Cuili Zhang
- Department of Environmental Health, School of Public Health, Shandong University, Jinan, China
| | - Jiliang Si
- Department of Environmental Health, School of Public Health, Shandong University, Jinan, China
| |
Collapse
|
30
|
Glosse P, Fajol A, Hirche F, Feger M, Voelkl J, Lang F, Stangl GI, Föller M. A high-fat diet stimulates fibroblast growth factor 23 formation in mice through TNFα upregulation. Nutr Diabetes 2018; 8:36. [PMID: 29807981 PMCID: PMC5972144 DOI: 10.1038/s41387-018-0037-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND/OBJECTIVES Bone-derived fibroblast growth factor 23 (FGF23) is a hormone that suppresses renal phosphate reabsorption and calcitriol (i.e., 1,25(OH)2D3) formation together with its co-receptor Klotho. FGF23- or Klotho-deficient mice suffer from rapid aging with multiple age-associated diseases, at least in part due to massive calcification. FGF23 is considered as a disease biomarker since elevated plasma levels are observed early in patients with acute and chronic disorders including renal, cardiovascular, inflammatory, and metabolic diseases. An energy-dense diet, which induces sequelae of the metabolic syndrome in humans and mice at least in part by enhancing pro-inflammatory TNFα formation, has recently been demonstrated to stimulate FGF23 production. METHODS We investigated the relevance of TNFα for high-fat diet (HFD)-induced FGF23 formation in wild-type (tnf+/+) and TNFα-deficient (tnf-/-) mice. RESULTS Within 3 weeks, HFD feeding resulted in a strong increase in the serum FGF23 level in tnf+/+ mice. Moreover, it caused low-grade inflammation as evident from a surge in hepatic Tnfα transcript levels. TNFα stimulated Fgf23 transcription in UMR106 osteoblast-like cells. Serum FGF23 was significantly lower in tnf-/- mice compared to tnf+/+ mice following HFD. Serum phosphate and calcitriol were not significantly affected by genotype or diet. CONCLUSIONS We show that HFD feeding is a powerful stimulator of murine FGF23 production through TNFα formation.
Collapse
Affiliation(s)
- Philipp Glosse
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120, Halle (Saale), Germany
| | - Abul Fajol
- Department of Physiology and Molecular Biology, Bangladesh University of Health Sciences (BUHS), Dhaka, 1216, Bangladesh.,Department of Physiology, University of Tübingen, 72076, Tübingen, Germany
| | - Frank Hirche
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120, Halle (Saale), Germany
| | - Martina Feger
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120, Halle (Saale), Germany
| | - Jakob Voelkl
- Department of Physiology, University of Tübingen, 72076, Tübingen, Germany
| | - Florian Lang
- Department of Physiology, University of Tübingen, 72076, Tübingen, Germany
| | - Gabriele I Stangl
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120, Halle (Saale), Germany
| | - Michael Föller
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120, Halle (Saale), Germany.
| |
Collapse
|
31
|
Zhang R, Li G, Zhang Q, Tang Q, Huang J, Hu C, Liu Y, Wang Q, Liu W, Gao N, Zhou S. Hirsutine induces mPTP-dependent apoptosis through ROCK1/PTEN/PI3K/GSK3β pathway in human lung cancer cells. Cell Death Dis 2018; 9:598. [PMID: 29789524 PMCID: PMC5964100 DOI: 10.1038/s41419-018-0641-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 04/13/2018] [Accepted: 05/02/2018] [Indexed: 02/02/2023]
Abstract
Hirsutine extracted from Uncaria rhynchophylla has been shown to exhibit anti-cancer activity. However, the molecular mechanism by which hirsutine exhibits anti-lung cancer activity remains unclear. In the present study, we showed that hirsutine induces apoptosis in human lung cancer cells via loss of mitochondrial membrane potential (∆ψm), adenosine triphosphate (ATP) depletion, ROS production, as well as cytochrome c release. Dephosphorylation of GSK3β is involved in hirsutine-mediated mitochondrial permeability transition pore (mPTP) opening through ANT1/CypD interaction. Mechanistic study revealed that interruption of ROCK1/PTEN/PI3K/Akt signaling pathway plays a critical role in hirsutine-mediated GSK3β dephosphorylation and mitochondrial apoptosis. Our in vivo study also showed that hirsutine effectively inhibits tumor growth in a A549 xenograft mouse model through ROCK1/PTEN/PI3K/Akt signaling-mediated GSK3β dephosphorylation and apoptosis. Collectively, these findings suggest a hierarchical model in which induction of apoptosis by hirsutine stems primarily from activation of ROCK1 and PTEN, inactivation of PI3K/Akt, leading in turn to GSK3β dephosphorylation and mPTP opening, and culminating in caspase-3 activation and apoptosis. These findings could provide a novel mechanistic basis for the application of hirsutine in the treatment of human lung cancer.
Collapse
Affiliation(s)
- Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 400037, Chongqing, China
| | - Guobing Li
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 400037, Chongqing, China
| | - Qian Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 400037, Chongqing, China
| | - Qin Tang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 400037, Chongqing, China
| | - Jingbin Huang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 400037, Chongqing, China
| | - Changpeng Hu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 400037, Chongqing, China
| | - Yali Liu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 400037, Chongqing, China
| | - Qing Wang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 400037, Chongqing, China
| | - Wuyi Liu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 400037, Chongqing, China
| | - Ning Gao
- College of Pharmacy, Army Medical University, 400038, Chongqing, China.
| | - Shiwen Zhou
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 400037, Chongqing, China.
| |
Collapse
|
32
|
Faulds KJ, Egelston JN, Sedivy LJ, Mitchell MK, Garimella S, Kozlowski H, D'Alessandro A, Hansen KC, Balsbaugh JL, Phiel CJ. Glycogen synthase kinase-3 (GSK-3) activity regulates mRNA methylation in mouse embryonic stem cells. J Biol Chem 2018; 293:10731-10743. [PMID: 29777057 DOI: 10.1074/jbc.ra117.001298] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/09/2018] [Indexed: 12/22/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) activity regulates multiple signal transduction pathways and is also a key component of the network responsible for maintaining stem cell pluripotency. Genetic deletion of Gsk-3α and Gsk-3β or inhibition of GSK-3 activity via small molecules promotes stem cell pluripotency, yet the mechanism underlying the role for GSK-3 in this process remains ambiguous. Another cellular process that has been shown to affect stem cell pluripotency is mRNA methylation (m6A). Here, we describe an intersection between these components, the regulation of m6A by GSK-3. We find that protein levels for the RNA demethylase, FTO (fat mass and obesity-associated protein), are elevated in Gsk-3α;Gsk-3β-deficient mouse embryonic stem cells (ESCs). FTO is normally phosphorylated by GSK-3, and MS identified the sites on FTO that are phosphorylated in a GSK-3-dependent fashion. GSK-3 phosphorylation of FTO leads to polyubiquitination, but in Gsk-3 knockout ESCs, that process is impaired, resulting in elevated levels of FTO protein. As a consequence of altered FTO protein levels, mRNAs in Gsk-3 knockout ESCs have 50% less m6A than WT ESCs, and m6A-Seq analysis reveals the specific mRNAs that have reduced m6A modifications. Taken together, we provide the first evidence for how m6A demethylation is regulated in mammalian cells and identify a putative novel mechanism by which GSK-3 activity regulates stem cell pluripotency.
Collapse
Affiliation(s)
- Kelsie J Faulds
- From the Department of Integrative Biology, University of Colorado Denver, Denver, Colorado 80204
| | - Jennifer N Egelston
- From the Department of Integrative Biology, University of Colorado Denver, Denver, Colorado 80204
| | - Laura J Sedivy
- From the Department of Integrative Biology, University of Colorado Denver, Denver, Colorado 80204
| | - Matthew K Mitchell
- From the Department of Integrative Biology, University of Colorado Denver, Denver, Colorado 80204
| | - Sanjana Garimella
- From the Department of Integrative Biology, University of Colorado Denver, Denver, Colorado 80204
| | - Hanna Kozlowski
- From the Department of Integrative Biology, University of Colorado Denver, Denver, Colorado 80204
| | - Angelo D'Alessandro
- the Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado 80045, and
| | - Kirk C Hansen
- the Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado 80045, and
| | - Jeremy L Balsbaugh
- the Mass Spectrometry Core Facility, Department of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, Colorado 80309
| | - Christopher J Phiel
- From the Department of Integrative Biology, University of Colorado Denver, Denver, Colorado 80204,
| |
Collapse
|
33
|
Insulin suppresses the production of fibroblast growth factor 23 (FGF23). Proc Natl Acad Sci U S A 2018; 115:5804-5809. [PMID: 29760049 DOI: 10.1073/pnas.1800160115] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factor 23 (FGF23) is produced by bone cells and regulates renal phosphate and vitamin D metabolism, as well as causing left ventricular hypertrophy. FGF23 deficiency results in rapid aging, whereas high plasma FGF23 levels are found in several disorders, including kidney or cardiovascular diseases. Regulators of FGF23 production include parathyroid hormone (PTH), calcitriol, dietary phosphate, and inflammation. We report that insulin and insulin-like growth factor 1 (IGF1) are negative regulators of FGF23 production. In UMR106 osteoblast-like cells, insulin and IGF1 down-regulated FGF23 production by inhibiting the transcription factor forkhead box protein O1 (FOXO1) through phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB)/Akt signaling. Insulin deficiency caused a surge in the serum FGF23 concentration in mice, which was reversed by administration of insulin. In women, a highly significant negative correlation between FGF23 plasma concentration and increase in plasma insulin level following an oral glucose load was found. Our results provide strong evidence that insulin/IGF1-dependent PI3K/PKB/Akt/FOXO1 signaling is a powerful suppressor of FGF23 production in vitro as well as in mice and in humans.
Collapse
|
34
|
Michael OS, Olatunji LA. Ameliorative effect of nicotine exposure on insulin resistance is accompanied by decreased cardiac glycogen synthase kinase-3 and plasminogen activator inhibitor-1 during oral oestrogen-progestin therapy. Arch Physiol Biochem 2018; 124:139-148. [PMID: 28868937 DOI: 10.1080/13813455.2017.1369549] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CONTEXT Cigarette smoking is considered to be a major risk factor for the development of diabetes and cardiovascular disease. Oestrogen-progestin combined oral contraceptive (COC) use has been associated with adverse cardiometabolic events. OBJECTIVE We hypothesized that nicotine would ameliorate insulin resistance (IR) that is accompanied by decreased cardiac glycogen synthase kinase-3 (GSK-3) and plasminogen activator inhibitor-1 (PAI-1). METHODS Female Wistar rats received (po) low-(0.1 mg/kg) or high-nicotine (1.0 mg/kg) with or without COC containing 5.0 µg levonorgestrel plus 1.0 µg ethinylestradiol daily for 8 weeks. RESULTS Data showed that COC treatment or nicotine exposure led to IR, glucose deregulation, atherogenic dyslipidemia, increased corticosterone, aldosterone, cardiac and circulating GSK-3 values and PAI-1. However, these effects with the exception of corticosterone and aldosterone were ameliorated in COC + nicotine-exposed rats. CONCLUSION Amelioration of IR induced by COC treatment is accompanied by decreased circulating PAI-1, cardiac PAI-1 and GSK-3 instead of circulating aldosterone and corticosterone.
Collapse
Affiliation(s)
- Olugbenga S Michael
- a Cardiovascular Research Laboratory, Department of Physiology , University of Ilorin, Ilorin, Nigeria
- b Hope Cardiometabolic Research Centre , Ilorin , Nigeria
- c Cardiometabolic Research Unit, Department of Physiology , College of Health sciences, Bowen University , Iwo , Nigeria
| | - Lawrence A Olatunji
- a Cardiovascular Research Laboratory, Department of Physiology , University of Ilorin, Ilorin, Nigeria
- b Hope Cardiometabolic Research Centre , Ilorin , Nigeria
| |
Collapse
|
35
|
Li J, Yu H, Wang S, Wang W, Chen Q, Ma Y, Zhang Y, Wang T. Natural products, an important resource for discovery of multitarget drugs and functional food for regulation of hepatic glucose metabolism. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:121-135. [PMID: 29391777 PMCID: PMC5768189 DOI: 10.2147/dddt.s151860] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Imbalanced hepatic glucose homeostasis is one of the critical pathologic events in the development of metabolic syndromes (MSs). Therefore, regulation of imbalanced hepatic glucose homeostasis is important in drug development for MS treatment. In this review, we discuss the major targets that regulate hepatic glucose homeostasis in human physiologic and pathophysiologic processes, involving hepatic glucose uptake, glycolysis and glycogen synthesis, and summarize their changes in MSs. Recent literature suggests the necessity of multitarget drugs in the management of MS disorder for regulation of imbalanced glucose homeostasis in both experimental models and MS patients. Here, we highlight the potential bioactive compounds from natural products with medicinal or health care values, and focus on polypharmacologic and multitarget natural products with effects on various signaling pathways in hepatic glucose metabolism. This review shows the advantage and feasibility of discovering multicompound-multitarget drugs from natural products, and providing a new perspective of ways on drug and functional food development for MSs.
Collapse
Affiliation(s)
- Jian Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin
| | - Haiyang Yu
- Department of Phytochemistry, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Sijian Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin
| | - Wei Wang
- Internal Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Qian Chen
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin
| | - Yanmin Ma
- Department of Phytochemistry, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin
| | - Tao Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin
| |
Collapse
|
36
|
Hermida MA, Dinesh Kumar J, Leslie NR. GSK3 and its interactions with the PI3K/AKT/mTOR signalling network. Adv Biol Regul 2017; 65:5-15. [PMID: 28712664 DOI: 10.1016/j.jbior.2017.06.003] [Citation(s) in RCA: 308] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 06/23/2017] [Indexed: 01/01/2023]
Abstract
Glycogen Synthase Kinase-3 (GSK3 or GSK-3) is a promiscuous protein kinase and its phosphorylation of its diverse substrates has major influences on many areas of physiology and pathology, including cellular metabolism, lineage commitment and neuroscience. GSK3 was one of the first identified substrates of the heavily studied oncogenic kinase AKT, phosphorylation by which inhibits GSK3 activity via the formation of an autoinhibitory pseudosubstrate sequence. This has led to investigation of the role of GSK3 inhibition as a key component of the cellular responses to growth factors and insulin, which stimulate the class I PI 3-Kinases and in turn AKT activity and GSK3 phosphorylation. GSK3 has been shown to phosphorylate several upstream and downstream components of the PI3K/AKT/mTOR signalling network, including AKT itself, RICTOR, TSC1 and 2, PTEN and IRS1 and 2, with the potential to apply feedback control within the network. However, it has been clear for some time that functionally distinct, insulated pools of GSK3 exist which are regulated independently, so that for some GSK3 substrates such as β-catenin, phosphorylation by GSK3 is not controlled by input from PI3K and AKT. Instead, as almost all GSK3 substrates require a priming phosphorylated residue to be 4 amino acids C-terminal to the Ser/Thr phosphorylated by GSK3, the predominant form of regulation of the activity of GSK3 often appears to be through control over these priming events, specific to individual substrates. Therefore, a major role of GSK3 can be viewed as an amplifier of the electrostatic effects on protein function which are caused by these priming phosphorylation events. Here we discuss these different aspects to GSK3 regulation and function, and the functions of GSK3 as it integrates with signalling through the PI3K-AKT-mTOR signalling axis.
Collapse
Affiliation(s)
- Miguel A Hermida
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot Watt University, Edinburgh EH14 4AS, UK
| | - J Dinesh Kumar
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot Watt University, Edinburgh EH14 4AS, UK
| | - Nick R Leslie
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot Watt University, Edinburgh EH14 4AS, UK.
| |
Collapse
|
37
|
Li Y, Yang P, Chang Q, Wang J, Liu J, Lv Y, Wang TTY, Gao B, Zhang Y, Yu LL. Inhibitory Effect of Piceatannol on TNF-α-Mediated Inflammation and Insulin Resistance in 3T3-L1 Adipocytes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:4634-4641. [PMID: 28535046 DOI: 10.1021/acs.jafc.7b01567] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Piceatannol, a bioactive component in grape and blueberry, was examined for its potential in decreasing the inflammatory activities in adipocytes using a cocultured adipocyte and macrophage system, and suppressing tumor necrosis factor-α (TNF-α)-mediated inflammation and the related insulin resistance using a 3T3-L1 adipocyte model. Piceatannol at 10 μM significantly reduced the release of inflammatory cytokines of TNF-α and monocyte chemoattractant protein-1 (MCP-1) by 19 and 31% in the cocultured system, respectively. Pretreatment with piceatannol also inhibited TNF-α-induced expression of interleukin-6 (IL-6) and MCP-1 at both mRNA and protein levels in the 3T3-L1 adipocytes. Piceatannol also partially improved the malfunction of insulin-stimulated glucose uptake, which was reduced by TNF-α in 3T3-L1 adipocytes. Furthermore, the inhibitions were mediated by significant blocking of IκBα phosphorylation and nuclear factor-κB (NF-κB) activation through suppressing nuclear translocation of NF-κB p65 along with c-Jun N-terminal kinase (JNK)-mitogen activated protein kinase (MAPK) activation. In addition, the Akt-dependent forkhead box O1 (FoxO1) signaling pathway was involved in the restoration of insulin-stimulated glucose uptake through suppressing the down-regulation of phosphorylation of Akt and FoxO1 expressions. These results suggested the potential of piceatannol in improving chronic inflammatory condition and insulin sensitivity in obese adipose tissues.
Collapse
Affiliation(s)
- Yanfang Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University , Beijing 100048, China
- Institute of Food and Nutraceutical Science, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Puyu Yang
- Institute of Food and Nutraceutical Science, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Qimeng Chang
- Department of Surgery, Fudan University Minhang Hospital , Shanghai 201199, China
| | - Jing Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University , Beijing 100048, China
| | - Jie Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University , Beijing 100048, China
| | - Yuan Lv
- Institute of Food and Nutraceutical Science, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Thomas T Y Wang
- Diet, Genomics, and Immunology Laboratory, Agricultural Research Service (ARS), U.S. Department of Agriculture , Beltsville, Maryland 20705, United States
| | - Boyan Gao
- Department of Nutrition and Food Science, University of Maryland , College Park, Maryland 20742, United States
| | - Yaqiong Zhang
- Institute of Food and Nutraceutical Science, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Liangli Lucy Yu
- Department of Nutrition and Food Science, University of Maryland , College Park, Maryland 20742, United States
| |
Collapse
|
38
|
Lin E, Kuo PH, Liu YL, Yang AC, Kao CF, Tsai SJ. Effects of circadian clock genes and health-related behavior on metabolic syndrome in a Taiwanese population: Evidence from association and interaction analysis. PLoS One 2017; 12:e0173861. [PMID: 28296937 PMCID: PMC5352001 DOI: 10.1371/journal.pone.0173861] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 02/28/2017] [Indexed: 12/12/2022] Open
Abstract
Increased risk of developing metabolic syndrome (MetS) has been associated with the circadian clock genes. In this study, we assessed whether 29 circadian clock-related genes (including ADCYAP1, ARNTL, ARNTL2, BHLHE40, CLOCK, CRY1, CRY2, CSNK1D, CSNK1E, GSK3B, HCRTR2, KLF10, NFIL3, NPAS2, NR1D1, NR1D2, PER1, PER2, PER3, REV1, RORA, RORB, RORC, SENP3, SERPINE1, TIMELESS, TIPIN, VIP, and VIPR2) are associated with MetS and its individual components independently and/or through complex interactions in a Taiwanese population. We also analyzed the interactions between environmental factors and these genes in influencing MetS and its individual components. A total of 3,000 Taiwanese subjects from the Taiwan Biobank were assessed in this study. Metabolic traits such as waist circumference, triglyceride, high-density lipoprotein cholesterol, systolic and diastolic blood pressure, and fasting glucose were measured. Our data showed a nominal association of MetS with several single nucleotide polymorphisms (SNPs) in five key circadian clock genes including ARNTL, GSK3B, PER3, RORA, and RORB; but none of these SNPs persisted significantly after performing Bonferroni correction. Moreover, we identified the effect of GSK3B rs2199503 on high fasting glucose (P = 0.0002). Additionally, we found interactions among the ARNTL rs10832020, GSK3B rs2199503, PER3 rs10746473, RORA rs8034880, and RORB rs972902 SNPs influenced MetS (P < 0.001 ~ P = 0.002). Finally, we investigated the influence of interactions between ARNTL rs10832020, GSK3B rs2199503, PER3 rs10746473, and RORB rs972902 with environmental factors such as alcohol consumption, smoking status, and physical activity on MetS and its individual components (P < 0.001 ~ P = 0.002). Our study indicates that circadian clock genes such as ARNTL, GSK3B, PER3, RORA, and RORB genes may contribute to the risk of MetS independently as well as through gene-gene and gene-environment interactions.
Collapse
Affiliation(s)
- Eugene Lin
- Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Vita Genomics, Inc., Taipei, Taiwan
- TickleFish Systems Corporation, Seattle, Western Australia, United States of America
- * E-mail: (EL); (SJT)
| | - Po-Hsiu Kuo
- Department of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Albert C. Yang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Psychiatry, National Yang-Ming University, Taipei, Taiwan
| | - Chung-Feng Kao
- Department of Agronomy, College of Agriculture & Natural Resources, National Chung Hsing University, Taichung, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Psychiatry, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (EL); (SJT)
| |
Collapse
|
39
|
Abstract
The decrease in adiponectin levels are negatively correlated with chronic subclinical inflammation markers in obesity. The hypertrophic adipocytes cause obesity-linked insulin resistance and metabolic syndrome. Furthermore, macrophage polarization is a key determinant regulating adiponectin receptor (AdipoR1/R2) expression and differential adiponectin-mediated macrophage inflammatory responses in obese individuals. In addition to decrease in adiponectin concentrations, the decline in AdipoR1/R2 mRNA expression leads to a decrement in adiponectin binding to cell membrane, and this turns into attenuation in the adiponectin effects. Within the receptor complex, adaptor protein-containing pleckstrin homology domain, phosphotyrosine-binding domain, and leucine zipper motif 1 (APPL1) is the intracellular binding partner of AdipoR1 and AdipoR2. The expression levels of APPL1 or APPL2 lead to an altered adiponectin activity. Despite normal or high adiponectin levels, an impaired post receptor signaling due to APPL1/APPL2 may alter adiponectin efficiency and activity. However, APPL2 blocks adiponectin signaling through AdipoR1 and AdipoR2 by competitive inhibition of APPL1. APPL1 is also an important mediator of adiponectin dependent insulin sensitization. In this context, adiponectin resistance is associated with insulin resistance and is thought to be partly due to the down-regulation of the AdipoRs in high-fat diet fed subjects. Actually, adiponectin resistance occurs very rapidly after saturated fatty acid feeding, this metabolic disturbance is not due to a decrease in AdipoR1 protein content. Intra-abdominal adipose tissue-AdipoR2 expression is reduced in obesity, whereas AdipoR1 expression is not changed. Adiponectin resistance together with insulin resistance forms a vicious cycle. The elevated adiponectin levels with adiponectin resistance is a compensatory response in the condition of an unusual discordance between insulin resistance and adiponectin unresponsiveness.Additionally, different mechanisms are involved in vascular adiponectin resistance at different stages of obesity. Nevertheless, diet-induced hyperlipidemia is the leading cause of vascular adiponectin resistance. Leptin/adiponectin imbalance may also be an important marker of the elevated risk of developing abdominal obesity-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- , Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|