1
|
Oh KK, Song SH, Park JH, Kim MJ, Kim DJ, Suk KT. A signpost to guide the key therapeutic components of Aralia continentalis Kitag roots in treating T2DM-derived heart attack, and diabetic nephropathy via systems biology concept. Life Sci 2025; 372:123635. [PMID: 40274257 DOI: 10.1016/j.lfs.2025.123635] [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: 12/22/2024] [Revised: 04/03/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
AIMS Aralia continentalis Kitag roots (ACKRs) have been regarded as a nutritional natural resource for treating different diseases, including type 2 diabetes mellitus (T2DM), and its complications (heart attack; HA, diabetic nephropathy; DN). Nonetheless, an extensive investigation of T2DM-derived complications has yet to be performed. MAIN METHODS Accordingly, we adopted gas chromatography-mass spectrometry (GC-MS) to identify the molecules of ACKRs, followed by the use of cheminformatics (Similarity Ensemble Approach; SEA, SwissTargetPrediction; STP), bioinformatics (STRING, DisGeNET, and OMIM), and computer screening tools to investigate its corresponding targets, in T2DM diseases and its complications. KEY FINDINGS The primary targets (PPARG, and IL6) were confirmed via a protein-protein interaction (PPI) network, suggesting that IL6- Andrographolide, PPARA-Germacrene D, PPARD- Kaurenoic acid, PPARG- Kaurenoic acid, NR1H3- 1-Naphthalenepropanol, α-ethenyldecahydro-5-(hydroxymethyl)-α,2,5,5,8a-pentamethyl-, and FABP4- Kaurenoic acid conformers on PPAR signaling pathway might exert agonistic mode. SIGNIFICANCE These findings underline that ACKRs' bioactives filtered by the devised platform could prevent T2DM-derived complications through multiple-target.
Collapse
Affiliation(s)
- Ki-Kwang Oh
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea.
| | - Seol Hee Song
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jeong Ha Park
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Min Ju Kim
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Dong Joon Kim
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Ki-Tae Suk
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea.
| |
Collapse
|
2
|
Talebi G, Saffarian P, Hakemi-Vala M, Sadeghi A, Yadegar A. The effect of Helicobacter pylori-derived extracellular vesicles on glucose metabolism and induction of insulin resistance in HepG2 cells. Arch Physiol Biochem 2025; 131:316-327. [PMID: 39431628 DOI: 10.1080/13813455.2024.2418494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/23/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
Helicobacter pylori infection has been associated with the development of insulin resistance (IR). This study aimed to examine the effect of H. pylori-derived extracellular vesicles (EVs) on IR induction. EVs were derived from two H. pylori strains, and characterised by transmission electron microscopy and dynamic light scattering. Different concentrations of insulin were added to HepG2 cells to induce IR model. HepG2 cells were exposed to various concentrations of H. pylori-derived EVs to assess IR development. The gene expression of IRS1, AKT2, GLUT2, IL-6, SOCS3, c-Jun and miR-140 was examined using RT-qPCR. Glucose uptake analysis revealed insulin at 5 × 10 -7 mol/l and EVs at 50 µg/ml induced IR model in HepG2 cells. H. pylori-derived EVs downregulated the expression level of IRS1, AKT2, and GLUT2, and upregulated IL-6, SOCS3, c-Jun, and miR-140 expression in HepG2 cells. In conclusion, our findings propose a novel mechanism by which H. pylori-derived EVs could potentially induce IR.
Collapse
Affiliation(s)
- Ghazaleh Talebi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Parvaneh Saffarian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mojdeh Hakemi-Vala
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Park MS, Hwang S, Kang HB, Ha M, Park J, Park SY, Park YJ, Park MH. Age-Dependent Effects of Butyl Benzyl Phthalate Exposure on Lipid Metabolism and Hepatic Fibrosis in Mice. Cells 2025; 14:126. [PMID: 39851554 PMCID: PMC11764096 DOI: 10.3390/cells14020126] [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: 12/23/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Endocrine-disrupting chemicals (EDCs), including phthalates, have been implicated in the development of non-alcoholic fatty liver disease (NAFLD) and hepatic fibrosis. This study investigates the age-dependent effects of butyl benzyl phthalate (BBP) exposure on lipid metabolism in the livers of young and aged mice. Young (2-month-old) and aged (20-month-old) male C57BL/6 mice were exposed to BBP through drinking water at a dose of 169 μg/kg/day for 6 and 4 months, respectively. Young mice exposed to BBP showed fatty liver, with downregulation of key fatty acid oxidation genes (CPT1A, CPT1B, CPT2, and Acox1) and elevated pro-inflammatory cytokines (TNF-α and IL-6). In contrast, aged mice exhibited hepatic fibrosis, with increased collagen deposition and upregulation of genes related to fibrosis (Acta2, MMP2, TGF-ß1, and Col1a2), cirrhosis (CXCR4, SOX9, DCN, and MFAP4), and cancer (Bcl2, CDKN2a, c-Myc, and Fn1). Overall, these findings emphasize the importance of age when evaluating the risks of EDC exposure, such as BBP. Future research should focus on understanding the molecular mechanisms behind these age-related differences and explore Grem1 and SOCS3 as potential therapeutic targets for treating EDC-induced and age-related liver diseases.
Collapse
Affiliation(s)
- Min-Seo Park
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (M.-S.P.); (S.H.); (H.-B.K.); (M.H.); (J.P.)
- Brain Busan 21 Plus Research Project Group, Kyungsung University, Busan 48434, Republic of Korea
| | - Seonhwa Hwang
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (M.-S.P.); (S.H.); (H.-B.K.); (M.H.); (J.P.)
- Brain Busan 21 Plus Research Project Group, Kyungsung University, Busan 48434, Republic of Korea
| | - Hyun-Bon Kang
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (M.-S.P.); (S.H.); (H.-B.K.); (M.H.); (J.P.)
| | - Minjeong Ha
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (M.-S.P.); (S.H.); (H.-B.K.); (M.H.); (J.P.)
| | - Juyeon Park
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (M.-S.P.); (S.H.); (H.-B.K.); (M.H.); (J.P.)
| | - So-Youn Park
- Department of Pharmaceutical Science and Technology, Kyungsung University, Busan 48434, Republic of Korea;
| | - Yong-Joo Park
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (M.-S.P.); (S.H.); (H.-B.K.); (M.H.); (J.P.)
- Brain Busan 21 Plus Research Project Group, Kyungsung University, Busan 48434, Republic of Korea
| | - Min-Hi Park
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (M.-S.P.); (S.H.); (H.-B.K.); (M.H.); (J.P.)
- Brain Busan 21 Plus Research Project Group, Kyungsung University, Busan 48434, Republic of Korea
| |
Collapse
|
4
|
Drosen ME, Bulbule S, Gottschalk G, Peterson D, Allen LA, Arnold LA, Roy A. Inactivation of ATG13 stimulates chronic demyelinating pathologies in muscle-serving nerves and spinal cord. Immunol Res 2025; 73:27. [PMID: 39777574 PMCID: PMC11706859 DOI: 10.1007/s12026-024-09557-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025]
Abstract
Chronic muscle fatigue is a condition characterized by debilitating muscle weakness and pain. Based on our recent finding to study the potential effect of mTOR on ATG13 inactivation in chronic muscle fatigue, we report that biweekly oral administration with MHY1485, a potent inducer of mTOR, develops chronic illness in mice resulting in severe muscle weakness. As a mechanism, we observed that MHY1485 feeding impaired ATG13-dependent autophagy, caused the infiltration of inflammatory M1 macrophages (Mφ), upregulated IL6 and RANTES by STAT3 activation, and augmented demyelination in muscle-serving nerve fibers. Interestingly, these mice displayed worsened muscle fatigue during 2-day post-treadmill exercise, suggesting the critical role of chronic mTOR activation in potential PEM pathogenesis. Interestingly, ATG13-repressor mice exhibited enhanced infiltration of M1Mφ cells, STAT3 activation, demyelination of nerve fibers, and PEM-like symptoms, suggesting the potential role of ATG13 impairment in post-exertional fatigue. HIGHLIGHTS: The potential role of mTOR activation in post-exertional fatigue is highlighted. As a molecular mechanism, mTOR activation augments autophagy impairment via ATG13 inactivation. Autophagy impairment induces IL-6 and RANTES via STAT3, demyelinates nerves in the muscle and spinal cord. ATG13 repressor mice (Tg-ATG13) displayed inflammatory demyelination and post-treadmill fatigue.
Collapse
Affiliation(s)
- Molly E Drosen
- Milwaukee Institute for Drug Discovery, Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI, 53211, USA
- Simmaron R&D Lab, 2000 E Kenwood Blvd, Suite # 320, Milwaukee, WI, 53211, USA
| | - Sarojini Bulbule
- Simmaron R&D Lab, 2000 E Kenwood Blvd, Suite # 320, Milwaukee, WI, 53211, USA
| | - Gunnar Gottschalk
- Simmaron R&D Lab, 2000 E Kenwood Blvd, Suite # 320, Milwaukee, WI, 53211, USA
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV, 89451, USA
| | | | - Linda Adrienne Allen
- Milwaukee Institute for Drug Discovery, Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI, 53211, USA
| | - Leggy A Arnold
- Milwaukee Institute for Drug Discovery, Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI, 53211, USA
| | - Avik Roy
- Milwaukee Institute for Drug Discovery, Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI, 53211, USA.
- Simmaron R&D Lab, 2000 E Kenwood Blvd, Suite # 320, Milwaukee, WI, 53211, USA.
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV, 89451, USA.
| |
Collapse
|
5
|
Mathews L, Appukuttan D, Victor DJ, Venkadassalapathy S, Subramanian S, Prakash PSG. Role of miRNA-155 in macrophage polarisation in stage III/IV periodontitis with type II diabetes mellitus: An analytical case-control study. Hum Immunol 2025; 86:111214. [PMID: 39667205 DOI: 10.1016/j.humimm.2024.111214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
AIM To evaluate the role of miR-155 in macrophage polarisation in stage III/IV periodontitis with Type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS Sixty four patients were recruited and categorized into Group I-systemically and Periodontally healthy (n = 16), Group II-systemically healthy with Stage III/IV Periodontitis(n = 16), Group III-Periodontally healthy with T2DM (n = 16) and Group IV- Stage III/IV Periodontitis with T2DM(n = 16).Gingival tissue samples were collected and Real time-PCR was carried out for microRNA-155, TNF- α(marker for M1 phenotype) and Arg-1(marker for M2 phenotype) gene expression. RESULTS Group IV showed the highest increase in miR-155 fold change (FC) (11.17 ± 10.44), followed by groups III and II(10.35 ± 15.87, 5.1 ± 5.17 respectively) when compared to Group I. Likewise, the highest FC for TNF-α expression was observed in the group IV (10.11 ± 12.14). Groups II and III showed an almost similar increase in the FC(4.36 ± 4.48 and 4.79 ± 6.91, respectively). Periodontally healthy subjects demonstrated higher levels of Arg-1 gene expression(4.41 ± 5.17), followed by reduced expression in the groups II and IV (2.46 ± 2.21 and 2.65 ± 3.25, respectively). The TNF-α:Arg-1 ratio indicated that group I had higher Arg-1 expression, while group III and IV individuals had higher TNF-α expression. Compared to miR-155 and Arg-1, which demonstrated poor Area under the curve(AUC), sensitivity and specificity, TNF-α was able to distinguish between the groups III & IV (AUC = 0.74, p = 0.02, sensitivity 92 %, specificity 83 %, cut off = 27.28) and I & IV (AUC = 0.6, p = 0.03, sensitivity 81 %, specificity 77 %, cut off = 26.04). CONCLUSION Dysregulated miR-155 contributes to hyperinflammatory state in T2DM associated periodontitis by favouring macrophage polarisation towards the M1 phenotype.However, the strength of this relationship and the association with severity of periodontal disease could not be confirmed in this study.
Collapse
Affiliation(s)
- Leya Mathews
- Postgraduate Student, Department of Periodontics, SRM Dental College, Bharathi Salai, Chennai, India.
| | - Devapriya Appukuttan
- Professor, Department of Periodontics, SRM Dental College, Bharathi Salai,Chennai, India.
| | - Dhayanand John Victor
- Professor and Head of the Department, Department of Periodontics, SRM Dental College, Bharathi Salai, Chennai, India
| | | | - Sangeetha Subramanian
- Professor, Department of Periodontics, SRM Dental College, Bharathi Salai,Chennai, India
| | - P S G Prakash
- Professor, Department of Periodontics, SRM Dental College, Bharathi Salai,Chennai, India
| |
Collapse
|
6
|
Jeremiah SS, Moin ASM, Butler AE. Virus-induced diabetes mellitus: revisiting infection etiology in light of SARS-CoV-2. Metabolism 2024; 156:155917. [PMID: 38642828 DOI: 10.1016/j.metabol.2024.155917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/14/2024] [Accepted: 04/14/2024] [Indexed: 04/22/2024]
Abstract
Diabetes mellitus (DM) is comprised of two predominant subtypes: type 1 diabetes mellitus (T1DM), accounting for approximately 5 % of cases worldwide and resulting from autoimmune destruction of insulin-producing β-cells, and type 2 (T2DM), accounting for approximately 95 % of cases globally and characterized by the inability of pancreatic β-cells to meet the demand for insulin due to a relative β-cell deficit in the setting of peripheral insulin resistance. Both types of DM involve derangement of glucose metabolism and are metabolic diseases generally considered to be initiated by a combination of genetic and environmental factors. Viruses have been reported to play a role as infectious etiological factors in the initiation of both types of DM in predisposed individuals. Among the reported viral infections causing DM in humans, the most studied include coxsackie B virus, cytomegalovirus and hepatitis C virus. The recent COVID-19 pandemic has highlighted the diabetogenic potential of SARS-CoV-2, rekindling interest in the field of virus-induced diabetes (VID). This review discusses the reported mechanisms of viral-induced DM, addressing emerging concepts in VID, as well as highlighting areas where knowledge is lacking, and further investigation is warranted.
Collapse
Affiliation(s)
| | - Abu Saleh Md Moin
- Royal College of Surgeons in Ireland - Medical University of Bahrain, Busaiteen, Kingdom of Bahrain.
| | - Alexandra E Butler
- Royal College of Surgeons in Ireland - Medical University of Bahrain, Busaiteen, Kingdom of Bahrain.
| |
Collapse
|
7
|
Šetinc M, Celinšćak Ž, Bočkor L, Zajc Petranović M, Stojanović Marković A, Peričić Salihović M, Deelen J, Škarić-Jurić T. The role of longevity-related genetic variant interactions as predictors of survival after 85 years of age. Mech Ageing Dev 2024; 219:111926. [PMID: 38484896 PMCID: PMC11166054 DOI: 10.1016/j.mad.2024.111926] [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: 01/26/2024] [Revised: 02/27/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024]
Abstract
Genome-wide association studies and candidate gene studies have identified several genetic variants that might play a role in achieving longevity. This study investigates interactions between pairs of those single nucleotide polymorphisms (SNPs) and their effect on survival above the age of 85 in a sample of 327 Croatian individuals. Although none of the SNPs individually showed a significant effect on survival in this sample, 14 of the 359 interactions tested (between SNPs not in LD) reached the level of nominal significance (p<0.05), showing a potential effect on late-life survival. Notably, SH2B3 rs3184504 interacted with different SNPs near TERC, TP53 rs1042522 with different SNPs located near the CDKN2B gene, and CDKN2B rs1333049 with different SNPs in FOXO3, as well as with LINC02227 rs2149954. The other interaction pairs with a possible effect on survival were FOXO3 rs2802292 and ERCC2 rs50871, IL6 rs1800795 and GHRHR rs2267723, LINC02227 rs2149954 and PARK7 rs225119, as well as PARK7 rs225119 and PTPN1 rs6067484. These interactions remained significant when tested together with a set of health-related variables that also had a significant effect on survival above 85 years. In conclusion, our results confirm the central role of genetic regulation of insulin signalling and cell cycle control in longevity.
Collapse
Affiliation(s)
- Maja Šetinc
- Institute for Anthropological Research, Zagreb 10000, Croatia; Centre for Applied Bioanthropology, Institute for Anthropological Research, Zagreb 10000, Croatia.
| | | | - Luka Bočkor
- Institute for Anthropological Research, Zagreb 10000, Croatia; Centre for Applied Bioanthropology, Institute for Anthropological Research, Zagreb 10000, Croatia
| | | | | | | | - Joris Deelen
- Max Planck Institute for Biology of Ageing, Cologne 50931, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany.
| | | |
Collapse
|
8
|
Chen C, Xie L, Zhang M, Shama, Cheng KKY, Jia W. The interplay between the muscle and liver in the regulation of glucolipid metabolism. J Mol Cell Biol 2024; 15:mjad073. [PMID: 38095440 PMCID: PMC11078061 DOI: 10.1093/jmcb/mjad073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/24/2023] [Indexed: 05/09/2024] Open
Affiliation(s)
- Cheng Chen
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Liping Xie
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Mingliang Zhang
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Shama
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Kenneth King Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Weiping Jia
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| |
Collapse
|
9
|
Lu H. Inflammatory liver diseases and susceptibility to sepsis. Clin Sci (Lond) 2024; 138:435-487. [PMID: 38571396 DOI: 10.1042/cs20230522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 01/09/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024]
Abstract
Patients with inflammatory liver diseases, particularly alcohol-associated liver disease and metabolic dysfunction-associated fatty liver disease (MAFLD), have higher incidence of infections and mortality rate due to sepsis. The current focus in the development of drugs for MAFLD is the resolution of non-alcoholic steatohepatitis and prevention of progression to cirrhosis. In patients with cirrhosis or alcoholic hepatitis, sepsis is a major cause of death. As the metabolic center and a key immune tissue, liver is the guardian, modifier, and target of sepsis. Septic patients with liver dysfunction have the highest mortality rate compared with other organ dysfunctions. In addition to maintaining metabolic homeostasis, the liver produces and secretes hepatokines and acute phase proteins (APPs) essential in tissue protection, immunomodulation, and coagulation. Inflammatory liver diseases cause profound metabolic disorder and impairment of energy metabolism, liver regeneration, and production/secretion of APPs and hepatokines. Herein, the author reviews the roles of (1) disorders in the metabolism of glucose, fatty acids, ketone bodies, and amino acids as well as the clearance of ammonia and lactate in the pathogenesis of inflammatory liver diseases and sepsis; (2) cytokines/chemokines in inflammatory liver diseases and sepsis; (3) APPs and hepatokines in the protection against tissue injury and infections; and (4) major nuclear receptors/signaling pathways underlying the metabolic disorders and tissue injuries as well as the major drug targets for inflammatory liver diseases and sepsis. Approaches that focus on the liver dysfunction and regeneration will not only treat inflammatory liver diseases but also prevent the development of severe infections and sepsis.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, U.S.A
| |
Collapse
|
10
|
Savage TM, Fortson KT, de Los Santos-Alexis K, Oliveras-Alsina A, Rouanne M, Rae SS, Gamarra JR, Shayya H, Kornberg A, Cavero R, Li F, Han A, Haeusler RA, Adam J, Schwabe RF, Arpaia N. Amphiregulin from regulatory T cells promotes liver fibrosis and insulin resistance in non-alcoholic steatohepatitis. Immunity 2024; 57:303-318.e6. [PMID: 38309273 PMCID: PMC10922825 DOI: 10.1016/j.immuni.2024.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/20/2023] [Accepted: 01/10/2024] [Indexed: 02/05/2024]
Abstract
Production of amphiregulin (Areg) by regulatory T (Treg) cells promotes repair after acute tissue injury. Here, we examined the function of Treg cells in non-alcoholic steatohepatitis (NASH), a setting of chronic liver injury. Areg-producing Treg cells were enriched in the livers of mice and humans with NASH. Deletion of Areg in Treg cells, but not in myeloid cells, reduced NASH-induced liver fibrosis. Chronic liver damage induced transcriptional changes associated with Treg cell activation. Mechanistically, Treg cell-derived Areg activated pro-fibrotic transcriptional programs in hepatic stellate cells via epidermal growth factor receptor (EGFR) signaling. Deletion of Areg in Treg cells protected mice from NASH-dependent glucose intolerance, which also was dependent on EGFR signaling on hepatic stellate cells. Areg from Treg cells promoted hepatocyte gluconeogenesis through hepatocyte detection of hepatic stellate cell-derived interleukin-6. Our findings reveal a maladaptive role for Treg cell-mediated tissue repair functions in chronic liver disease and link liver damage to NASH-dependent glucose intolerance.
Collapse
Affiliation(s)
- Thomas M Savage
- Department of Microbiology & Immunology, Columbia University, New York, NY, USA
| | - Katherine T Fortson
- Department of Microbiology & Immunology, Columbia University, New York, NY, USA
| | | | | | - Mathieu Rouanne
- Department of Microbiology & Immunology, Columbia University, New York, NY, USA
| | - Sarah S Rae
- Department of Microbiology & Immunology, Columbia University, New York, NY, USA
| | | | - Hani Shayya
- Mortimer B. Zuckerman Mind, and Brain and Behavior Institute, Columbia University, New York, NY, USA
| | - Adam Kornberg
- Department of Microbiology & Immunology, Columbia University, New York, NY, USA; Columbia Center for Translational Immunology, Columbia University, New York, NY, USA
| | - Renzo Cavero
- Department of Microbiology & Immunology, Columbia University, New York, NY, USA
| | - Fangda Li
- Department of Microbiology & Immunology, Columbia University, New York, NY, USA
| | - Arnold Han
- Department of Microbiology & Immunology, Columbia University, New York, NY, USA; Columbia Center for Translational Immunology, Columbia University, New York, NY, USA; Department of Medicine, Columbia University, New York, NY, USA
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Julien Adam
- Pathology Department, Hopital Paris Saint-Joseph, Paris, France; INSERM U1186, Gustave Roussy, Villejuif, France
| | | | - Nicholas Arpaia
- Department of Microbiology & Immunology, Columbia University, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA.
| |
Collapse
|
11
|
Engin A. Protein Kinases in Obesity, and the Kinase-Targeted Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:199-229. [PMID: 39287853 DOI: 10.1007/978-3-031-63657-8_7] [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 action of protein kinases and protein phosphatases is essential for multiple physiological responses. Each protein kinase displays its own unique substrate specificity and a regulatory mechanism that may be modulated by association with other proteins. Protein kinases are classified as dual-specificity kinases and dual-specificity phosphatases. Dual-specificity phosphatases are important signal transduction enzymes that regulate various cellular processes in coordination with protein kinases and play an important role in obesity. Impairment of insulin signaling in obesity is largely mediated by the activation of the inhibitor of kappa B-kinase beta and the c-Jun N-terminal kinase (JNK). Oxidative stress and endoplasmic reticulum (ER) stress activate the JNK pathway which suppresses insulin biosynthesis. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) are important for proper regulation of glucose metabolism in mammals at both the hormonal and cellular levels. Additionally, obesity-activated calcium/calmodulin dependent-protein kinase II/p38 suppresses insulin-induced protein kinase B phosphorylation by activating the ER stress effector, activating transcription factor-4. To alleviate lipotoxicity and insulin resistance, promising targets are pharmacologically inhibited. Nifedipine, calcium channel blocker, stimulates lipogenesis and adipogenesis by downregulating AMPK and upregulating mTOR, which thereby enhances lipid storage. Contrary to the nifedipine, metformin activates AMPK, increases fatty acid oxidation, suppresses fatty acid synthesis and deposition, and thus alleviates lipotoxicity. Obese adults with vascular endothelial dysfunction have greater endothelial cells activation of unfolded protein response stress sensors, RNA-dependent protein kinase-like ER eukaryotic initiation factor-2 alpha kinase (PERK), and activating transcription factor-6. The transcriptional regulation of adipogenesis in obesity is influenced by AGC (protein kinase A (PKA), PKG, PKC) family signaling kinases. Obesity may induce systemic oxidative stress and increase reactive oxygen species in adipocytes. An increase in intracellular oxidative stress can promote PKC-β activation. Activated PKC-β induces growth factor adapter Shc phosphorylation. Shc-generated peroxides reduce mitochondrial oxygen consumption and enhance triglyceride accumulation and lipotoxicity. Liraglutide attenuates mitochondrial dysfunction and reactive oxygen species generation. Co-treatment of antiobesity and antidiabetic herbal compound, berberine with antipsychotic drug olanzapine decreases the accumulation of triglyceride. While low-dose rapamycin, metformin, amlexanox, thiazolidinediones, and saroglitazar protect against insulin resistance, glucagon-like peptide-1 analog liraglutide inhibits palmitate-induced inflammation by suppressing mTOR complex 1 (mTORC1) activity and protects against lipotoxicity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
12
|
Arévalo J, Campoy I, Durán M, Nemours S, Areny A, Vall-Palomar M, Martínez C, Cantero-Recasens G, Meseguer A. STAT3 phosphorylation at serine 727 activates specific genetic programs and promotes clear cell renal cell carcinoma (ccRCC) aggressiveness. Sci Rep 2023; 13:19552. [PMID: 37945711 PMCID: PMC10636117 DOI: 10.1038/s41598-023-46628-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023] Open
Abstract
The signal transducer and activator of transcription 3 (STAT3) is a transcription factor mainly activated by phosphorylation in either tyrosine 705 (Y705) or serine 727 (S727) residues that regulates essential processes such as cell differentiation, apoptosis inhibition, or cell survival. Aberrant activation of STAT3 has been related to development of nearly 50% of human cancers including clear cell renal cell carcinoma (ccRCC). In fact, phosho-S727 (pS727) levels correlate with overall survival of ccRCC patients. With the aim to elucidate the contribution of STAT3 phosphorylation in ccRCC development and progression, we have generated human-derived ccRCC cell lines carrying STAT3 Y705 and S727 phosphomutants. Our data show that the phosphomimetic substitution Ser727Asp facilitates a pro-tumoral phenotype in vitro, in a Y705-phosphorylation-independent manner. Moreover, we describe that STAT3 phosphorylation state determines the expression of different subsets of target genes associated with distinct biological processes, being pS727-dependent genes the most related to cellular hallmarks of cancer. In summary, the present study constitutes the first analysis on the role of overall STAT3 phosphorylation state in ccRCC and demonstrates that pS727 promotes the expression of a specific subset of target genes that might be clinically relevant as novel biomarkers and potential therapeutic targets for ccRCC.
Collapse
Affiliation(s)
- J Arévalo
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| | - I Campoy
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - M Durán
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - S Nemours
- Molecular Oncology Group, Biodonostia Health Research Institute, Paseo Dr. Begiristain, s/n, 20014, San Sebastián, Spain
| | - A Areny
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - M Vall-Palomar
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - C Martínez
- Vascular and Renal Translational Research Group, Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198, Lleida, Spain
| | - G Cantero-Recasens
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - A Meseguer
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Unitat de Bioquímica de Medicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
13
|
Orange ST, Leslie J, Ross M, Mann DA, Wackerhage H. The exercise IL-6 enigma in cancer. Trends Endocrinol Metab 2023; 34:749-763. [PMID: 37633799 DOI: 10.1016/j.tem.2023.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/28/2023]
Abstract
Interleukin (IL)-6 elicits both anticancer and procancer effects depending on the context, which we have termed the 'exercise IL-6 enigma'. IL-6 is released from skeletal muscles during exercise to regulate short-term energy availability. Exercise-induced IL-6 provokes biological effects that may protect against cancer by improving insulin sensitivity, stimulating the production of anti-inflammatory cytokines, mobilising immune cells, and reducing DNA damage in early malignant cells. By contrast, IL-6 continuously produced by leukocytes in inflammatory sites drives tumorigenesis by promoting chronic inflammation and activating tumour-promoting signalling pathways. How can a molecule have such opposing effects on cancer? Here, we review the roles of IL-6 in chronic inflammation, tumorigenesis, and exercise-associated cancer prevention and define the factors that underpin the exercise IL-6 enigma.
Collapse
Affiliation(s)
- Samuel T Orange
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; School of Biomedical, Nutritional and Sport Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| | - Jack Leslie
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mark Ross
- Institute of Life and Earth Sciences, School of Energy, Geoscience, Infrastructure and Society, Heriot-Watt University, Edinburgh, UK
| | - Derek A Mann
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Henning Wackerhage
- Department of Sport & Health Science, Technical University of Munich, Munich, Germany
| |
Collapse
|
14
|
Lim JY, Kim E. The Role of Organokines in Obesity and Type 2 Diabetes and Their Functions as Molecular Transducers of Nutrition and Exercise. Metabolites 2023; 13:979. [PMID: 37755259 PMCID: PMC10537761 DOI: 10.3390/metabo13090979] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023] Open
Abstract
Maintaining systemic homeostasis requires the coordination of different organs and tissues in the body. Our bodies rely on complex inter-organ communications to adapt to perturbations or changes in metabolic homeostasis. Consequently, the liver, muscle, and adipose tissues produce and secrete specific organokines such as hepatokines, myokines, and adipokines in response to nutritional and environmental stimuli. Emerging evidence suggests that dysregulation of the interplay of organokines between organs is associated with the pathophysiology of obesity and type 2 diabetes (T2D). Strategies aimed at remodeling organokines may be effective therapeutic interventions. Diet modification and exercise have been established as the first-line therapeutic intervention to prevent or treat metabolic diseases. This review summarizes the current knowledge on organokines secreted by the liver, muscle, and adipose tissues in obesity and T2D. Additionally, we highlighted the effects of diet/nutrition and exercise on the remodeling of organokines in obesity and T2D. Specifically, we investigated the ameliorative effects of caloric restriction, selective nutrients including ω3 PUFAs, selenium, vitamins, and metabolites of vitamins, and acute/chronic exercise on the dysregulation of organokines in obesity and T2D. Finally, this study dissected the underlying molecular mechanisms by which nutrition and exercise regulate the expression and secretion of organokines in specific tissues.
Collapse
Affiliation(s)
- Ji Ye Lim
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), 6431 Fannin St., Houston, TX 77030, USA
| | - Eunju Kim
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), 6431 Fannin St., Houston, TX 77030, USA
| |
Collapse
|
15
|
Cao R, Tian H, Zhang Y, Liu G, Xu H, Rao G, Tian Y, Fu X. Signaling pathways and intervention for therapy of type 2 diabetes mellitus. MedComm (Beijing) 2023; 4:e283. [PMID: 37303813 PMCID: PMC10248034 DOI: 10.1002/mco2.283] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/18/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) represents one of the fastest growing epidemic metabolic disorders worldwide and is a strong contributor for a broad range of comorbidities, including vascular, visual, neurological, kidney, and liver diseases. Moreover, recent data suggest a mutual interplay between T2DM and Corona Virus Disease 2019 (COVID-19). T2DM is characterized by insulin resistance (IR) and pancreatic β cell dysfunction. Pioneering discoveries throughout the past few decades have established notable links between signaling pathways and T2DM pathogenesis and therapy. Importantly, a number of signaling pathways substantially control the advancement of core pathological changes in T2DM, including IR and β cell dysfunction, as well as additional pathogenic disturbances. Accordingly, an improved understanding of these signaling pathways sheds light on tractable targets and strategies for developing and repurposing critical therapies to treat T2DM and its complications. In this review, we provide a brief overview of the history of T2DM and signaling pathways, and offer a systematic update on the role and mechanism of key signaling pathways underlying the onset, development, and progression of T2DM. In this content, we also summarize current therapeutic drugs/agents associated with signaling pathways for the treatment of T2DM and its complications, and discuss some implications and directions to the future of this field.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Huimin Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yu Zhang
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Geng Liu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Haixia Xu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Guocheng Rao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yan Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Xianghui Fu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
16
|
Xu H, Zhao Y, Zhao Q, Shi M, Zhang Z, Ding W, Zhao Y. Tuberous Sclerosis Complex 1 Deficiency in Macrophages Promotes Unclassical Inflammatory Response to Lipopolysaccharide In Vitro and Dextran Sodium Sulfate-Induced Colitis in Mice. Aging Dis 2022; 13:1875-1890. [PMID: 36465179 PMCID: PMC9662278 DOI: 10.14336/ad.2022.0408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 04/08/2022] [Indexed: 09/01/2023] Open
Abstract
Human tuberous sclerosis (TSC) is mainly caused by genetic mutations of tuberous TSC1or TSC2. Recent studies found that TSC1 deficiency promoted classical M1 macrophage polarization. However, whether TSC1 regulates other inflammatory cytokine expression in lipopolysaccharidem (LPS)-stimulated macrophages is unknown. Herein, we studied the cytokine expression profile of wild-type (WT) and TSC1-deleted macrophages after LPS stimulation in vitro and the pathogenesis of dextran sodium sulfate (DSS)-induced colitis in mice with myeloid-specific TSC1 deletion (TSC1cKO mice). We found that TSC1-deficient macrophages exhibited the enhanced secretion of interleukin-17A (IL-17A), IL-17F, and interferon-gamma (IFN-γ) in response to LPS stimulation in vitro. This is in contrast to LPS-stimulated WT macrophages, which usually do not. Importantly, TSC1cKO mice exhibited exacerbated DSS-induced acute colitis with severer symptoms. MTOR deletion or rapamycin treatment significantly reversed the enhanced expressions of IL-17A, IL-17F, and IFN-γ in LPS-stimulated TSC1-deficient macrophages in vitro and rescued the enhanced DSS-induced colitis in TSC1cKO mice, indicating that TSC1 deficiency increased these cytokine productions in an mTOR-dependent manner. RNA-sequencing and molecular studies indicated that TSC1 deficiency enhanced the aerobic glycolysis process and the activities of mTOR-STAT3-RORγT pathway in LPS-stimulated macrophages. Inhibition of aerobic glycolysis, STAT3, or RORγT reversed IL-17 and IFN-γ expression in LPS-treated TSC1-deficient macrophages. Thus, TSC1 is essential for macrophages to shut down IL-17A, IL-17F, and IFN-γ expression during LPS stimulation by suppressing the aerobic glycolysis process and mTOR-STAT3, RORγT, and T-bet pathways. The present study uncovered the key role of TSC1 in shutting down IL-17A, IL-17F, and IFN-γ expressions in LPS-treated macrophages.
Collapse
Affiliation(s)
- Huawen Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Cunji Medical College, University of Chinese Academy of Sciences, Beijing, China.
| | - Yang Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Cunji Medical College, University of Chinese Academy of Sciences, Beijing, China.
| | - Qingjie Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
| | - Mingpu Shi
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Cunji Medical College, University of Chinese Academy of Sciences, Beijing, China.
| | - Zhaoqi Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Cunji Medical College, University of Chinese Academy of Sciences, Beijing, China.
| | - Wenjun Ding
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Cunji Medical College, University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
17
|
Serum Interleukins as Potential Prognostic Biomarkers in HBV-Related Acute-on-Chronic Liver Failure. Mediators Inflamm 2022; 2022:7794890. [PMID: 36117587 PMCID: PMC9477565 DOI: 10.1155/2022/7794890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/06/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatitis B virus-related acute-on-chronic liver failure (HBV-ACLF) is relatively common in China and has complex pathogenesis, difficult clinical treatment, and poor prognosis. Immune status is an important factor affecting ACLF prognosis. Interleukins are a family of secreted lymphocyte factors that interact with a host of cell types including immune cells. These signaling molecules play important roles in transmitting information; regulating immune cells; mediating the activation, proliferation, and differentiation of T and B cells; and modulating inflammatory responses. Many studies have investigated the correlation between interleukin expression and the prognosis of HBV-ACLF. This review focuses on the potential use of interleukins as prognostic biomarkers in HBV-ACLF. References were mainly identified through PubMed and CNKI search, including relevant studies published until December 2021. We have summarized reports of several promising diagnostic interleukin biomarkers that predict susceptibility to HBV-ACLF. The use of biomarkers to understand early prognosis can help devise different therapeutic measures and improve patient survival. Ongoing research on prognostic biomarkers of HBV-ACLF is promising, and future preclinical and clinical studies are warranted.
Collapse
|
18
|
Tao Y, Jiang Q, Wang Q. Adipose tissue macrophages in remote modulation of hepatic glucose production. Front Immunol 2022; 13:998947. [PMID: 36091076 PMCID: PMC9449693 DOI: 10.3389/fimmu.2022.998947] [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: 07/20/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Hepatic glucose production (HGP) is fine-regulated via glycogenolysis or gluconeogenesis to maintain physiological concentration of blood glucose during fasting-feeding cycle. Aberrant HGP leads to hyperglycemia in obesity-associated diabetes. Adipose tissue cooperates with the liver to regulate glycolipid metabolism. During these processes, adipose tissue macrophages (ATMs) change their profiles with various physio-pathological settings, producing diverse effects on HGP. Here, we briefly review the distinct phenotypes of ATMs under different nutrition states including feeding, fasting or overnutrition, and detail their effects on HGP. We discuss several pathways by which ATMs regulate hepatic gluconeogenesis or glycogenolysis, leading to favorable or unfavorable metabolic consequences. Furthermore, we summarize emerging therapeutic targets to correct metabolic disorders in morbid obesity or diabetes based on ATM-HGP axis. This review puts forward the importance and flexibility of ATMs in regulating HGP, proposing ATM-based HGP modulation as a potential therapeutic approach for obesity-associated metabolic dysfunction.
Collapse
Affiliation(s)
| | | | - Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
19
|
Pinto AP, Rocha ALD, Marafon BB, Nogueira JE, Branco LGS, Pauli JR, de Moura LP, Cintra DE, Ropelle ER, da Silva ASR. Chronic rapamycin treatment decreases hepatic
IL
‐6 protein but increases autophagy markers as a protective effect against the overtraining‐induced tissue damage. Clin Exp Pharmacol Physiol 2022; 49:893-902. [PMID: 35637552 DOI: 10.1111/1440-1681.13677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/13/2022] [Accepted: 05/25/2022] [Indexed: 01/18/2023]
Affiliation(s)
- Ana P. Pinto
- Postgraduate Program in Rehabilitation and Functional Performance Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto São Paulo Brazil
| | - Alisson L. da Rocha
- Postgraduate Program in Rehabilitation and Functional Performance Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto São Paulo Brazil
| | - Bruno B. Marafon
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto São Paulo Brazil
| | - Jonatas E. Nogueira
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto São Paulo Brazil
| | - Luiz G. S. Branco
- Department of Basic and Oral Biology, Dental School of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| | - José R. Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx) School of Applied Sciences, University of Campinas (UNICAMP), Limeira São Paulo Brazil
| | - Leandro P. de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx) School of Applied Sciences, University of Campinas (UNICAMP), Limeira São Paulo Brazil
| | - Dennys E. Cintra
- Laboratory of Molecular Biology of Exercise (LaBMEx) School of Applied Sciences, University of Campinas (UNICAMP), Limeira São Paulo Brazil
| | - Eduardo R. Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx) School of Applied Sciences, University of Campinas (UNICAMP), Limeira São Paulo Brazil
| | - Adelino S. R. da Silva
- Postgraduate Program in Rehabilitation and Functional Performance Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto São Paulo Brazil
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto São Paulo Brazil
| |
Collapse
|
20
|
Ding Q, Gao Z, Chen K, Zhang Q, Hu S, Zhao L. Inflammation-Related Epigenetic Modification: The Bridge Between Immune and Metabolism in Type 2 Diabetes. Front Immunol 2022; 13:883410. [PMID: 35603204 PMCID: PMC9120428 DOI: 10.3389/fimmu.2022.883410] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/11/2022] [Indexed: 12/11/2022] Open
Abstract
T2DM, as a typical metabolic inflammatory disease, is under the joint regulation of environmental factors and genetics, combining with a variety of epigenetic changes. Apart from epigenetic changes of islet β cells and glycometabolic tissues or organs, the inflammation-related epigenetics is also the core pathomechanism leading to β-cell dysfunction and insulin resistance. In this review, we focus on the epigenetic modification of immune cells’ proliferation, recruitment, differentiation and function, providing an overview of the key genes which regulated by DNA methylation, histone modifications, and non-coding RNA in the respect of T2DM. Meanwhile, we further summarize the present situation of T2DM epigenetic research and elucidate its prospect in T2DM clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Qiyou Ding
- Department of Endocrinology, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zezheng Gao
- Department of Endocrinology, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Keyu Chen
- Department of Endocrinology, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Qiqi Zhang
- Department of Endocrinology, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shiwan Hu
- Department of Endocrinology, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Linhua Zhao,
| |
Collapse
|
21
|
Antuna-Puente B, Fellahi S, McAvoy C, Fève B, Bastard JP. Interleukins in adipose tissue: Keeping the balance. Mol Cell Endocrinol 2022; 542:111531. [PMID: 34910978 DOI: 10.1016/j.mce.2021.111531] [Citation(s) in RCA: 6] [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] [Received: 04/26/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023]
Abstract
The role of the immune system is to defend the host and preserve the functionality in response to stress. This function is not limited to infection or injury as it also plays a role in the response to overnutrition. Indeed, low-grade chronic activation of the immune system associated with overnutrition may be deleterious, contributing importantly to diabetes and long-term complications, such as cardiovascular disorders. Increasing evidence shows that adipose tissue participates in the obesity-related inflammatory response and that interleukins are one of the key players, either as a pro-inflammatory response to the metabolic dysregulation or to restore homeostasis. The crosstalk between adipocytes and immune cells through some important interleukins and their role in metabolic disruption is the topic of this review.
Collapse
Affiliation(s)
- Barbara Antuna-Puente
- Infection Disease Division, Department of Medicine, Queen's University, Kingston, ON, Canada.
| | - Soraya Fellahi
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Département de Biochimie-pharmacologie-biologie Moléculaire-génétique Médicale, Créteil, France; Sorbonne Université-Inserm, Centre de Recherche Saint-Antoine UMR S_938, 75012, Paris Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Paris, France
| | - Chloé McAvoy
- Unité de Recherche Clinique de L'Est Parisien (URC-Est), Hôpital Saint Antoine, Paris, France
| | - Bruno Fève
- Sorbonne Université-Inserm, Centre de Recherche Saint-Antoine UMR S_938, 75012, Paris Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Paris, France; Assistance Publique- Hôpitaux de Paris -Hôpital Saint-Antoine, Service D'Endocrinologie-Diabétologie, Centre de Référence des Maladies Rares de L'Insulino-Sécrétion et de L'Insulino-Sensibilité (PRISIS), 75012, Paris, France
| | - Jean-Philippe Bastard
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Département de Biochimie-pharmacologie-biologie Moléculaire-génétique Médicale, Créteil, France; FHU-SENEC, INSERM U955 and Université Paris Est (UPEC), UMR U955, Faculté de Santé, Créteil, France
| |
Collapse
|
22
|
de Mendonça ELSS, Fragoso MBT, de Oliveira JM, Xavier JA, Goulart MOF, de Oliveira ACM. Gestational Diabetes Mellitus: The Crosslink among Inflammation, Nitroxidative Stress, Intestinal Microbiota and Alternative Therapies. Antioxidants (Basel) 2022; 11:129. [PMID: 35052633 PMCID: PMC8773111 DOI: 10.3390/antiox11010129] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 01/09/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is characterized by a set of metabolic complications arising from adaptive failures to the pregnancy period. Estimates point to a prevalence of 3 to 15% of pregnancies. Its etiology includes intrinsic and extrinsic aspects of the progenitress, which may contribute to the pathophysiogenesis of GDM. Recently, researchers have identified that inflammation, oxidative stress, and the gut microbiota participate in the development of the disease, with potentially harmful effects on the health of the maternal-fetal binomial, in the short and long terms. In this context, alternative therapies were investigated from two perspectives: the modulation of the intestinal microbiota, with probiotics and prebiotics, and the use of natural products with antioxidant and anti-inflammatory properties, which may mitigate the endogenous processes of the GDM, favoring the health of the mother and her offspring, and in a future perspective, alleviating this critical public health problem.
Collapse
Affiliation(s)
- Elaine Luiza Santos Soares de Mendonça
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceio 57072-970, Alagoas, Brazil; (E.L.S.S.d.M.); (M.B.T.F.); (J.M.d.O.); (J.A.X.)
| | - Marilene Brandão Tenório Fragoso
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceio 57072-970, Alagoas, Brazil; (E.L.S.S.d.M.); (M.B.T.F.); (J.M.d.O.); (J.A.X.)
| | - Jerusa Maria de Oliveira
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceio 57072-970, Alagoas, Brazil; (E.L.S.S.d.M.); (M.B.T.F.); (J.M.d.O.); (J.A.X.)
| | - Jadriane Almeida Xavier
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceio 57072-970, Alagoas, Brazil; (E.L.S.S.d.M.); (M.B.T.F.); (J.M.d.O.); (J.A.X.)
| | - Marília Oliveira Fonseca Goulart
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceio 57072-970, Alagoas, Brazil; (E.L.S.S.d.M.); (M.B.T.F.); (J.M.d.O.); (J.A.X.)
| | | |
Collapse
|
23
|
Greenbaum CJ, Serti E, Lambert K, Weiner LJ, Kanaparthi S, Lord S, Gitelman SE, Wilson DM, Gaglia JL, Griffin KJ, Russell WE, Raskin P, Moran A, Willi SM, Tsalikian E, DiMeglio LA, Herold KC, Moore WV, Goland R, Harris M, Craig ME, Schatz DA, Baidal DA, Rodriguez H, Utzschneider KM, Nel HJ, Soppe CL, Boyle KD, Cerosaletti K, Keyes-Elstein L, Long SA, Thomas R, McNamara JG, Buckner JH, Sanda S. IL-6 receptor blockade does not slow β cell loss in new-onset type 1 diabetes. JCI Insight 2021; 6:150074. [PMID: 34747368 PMCID: PMC8663550 DOI: 10.1172/jci.insight.150074] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/22/2021] [Indexed: 12/30/2022] Open
Abstract
BackgroundIL-6 receptor (IL-6R) signaling drives development of T cell populations important to type 1 diabetes pathogenesis. We evaluated whether blockade of IL-6R with monoclonal antibody tocilizumab would slow loss of residual β cell function in newly diagnosed type 1 diabetes patients.MethodsWe conducted a multicenter, randomized, placebo-controlled, double-blind trial with tocilizumab in new-onset type 1 diabetes. Participants were screened within 100 days of diagnosis. Eligible participants were randomized 2:1 to receive 7 monthly doses of tocilizumab or placebo. The primary outcome was the change from screening in the mean AUC of C-peptide collected during the first 2 hours of a mixed meal tolerance test at week 52 in pediatric participants (ages 6-17 years).ResultsThere was no statistical difference in the primary outcome between tocilizumab and placebo. Immunophenotyping showed reductions in downstream signaling of the IL-6R in T cells but no changes in CD4 memory subsets, Th17 cells, Tregs, or CD4+ T effector cell resistance to Treg suppression. A DC subset decreased during therapy but regressed to baseline once therapy stopped. Tocilizumab was well tolerated.ConclusionTocilizumab reduced T cell IL-6R signaling but did not modulate CD4+ T cell phenotypes or slow loss of residual β cell function in newly diagnosed individuals with type 1 diabetes.Trial RegistrationClinicalTrials.gov NCT02293837.FundingNIH National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) and National Institute of Allergy and Infectious Diseases (NIAID) UM1AI109565, UL1TR000004 from NIH/National Center for Research Resources (NCRR) Clinical and Translational Science Award (CTSA), NIH/NIDDK P30DK036836, NIH/NIDDK U01DK103266, NIH/NIDDK U01DK103266, 1UL1TR000064 from NIH/NCRR CTSA, NIH/National Center for Advancing Translational Sciences (NCATS) UL1TR001878, UL1TR002537 from NIH/CTSA; National Health and Medical Research Council Practitioner Fellowship (APP1136735), NIH/NIDDK U01-DK085476, NIH/CTSA UL1-TR002494, Indiana Clinical and Translational Science Institute Award UL1TR002529, Vanderbilt Institute for Clinical and Translational Research UL1TR000445. NIH/NCATS UL1TR003142, NIH/CTSA program UL1-TR002494, Veteran Affairs Administration, and 1R01AI132774.
Collapse
Affiliation(s)
- Carla J Greenbaum
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, Washington, USA
| | | | - Katharina Lambert
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, Washington, USA
| | | | | | - Sandra Lord
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, Washington, USA
| | | | | | - Jason L Gaglia
- Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Philip Raskin
- University of Texas, Southwestern, Dallas, Texas, USA
| | | | - Steven M Willi
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Linda A DiMeglio
- Riley Children's Hospital, Indiana University, Indianapolis, Indiana, USA
| | | | - Wayne V Moore
- University of Missouri, Kansas City, Kansas City, Missouri, USA
| | | | - Mark Harris
- Children's Health Queensland Hospital, South Brisbane, Australia.,University of Queensland, Queensland, Brisbane, Australia
| | - Maria E Craig
- University of Sydney, Sydney New South Wales, Australia
| | | | | | | | | | - Hendrik J Nel
- University of Queensland, Queensland, Brisbane, Australia
| | | | | | - Karen Cerosaletti
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, Washington, USA
| | | | - S Alice Long
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, Washington, USA
| | - Ranjeny Thomas
- University of Queensland, Queensland, Brisbane, Australia
| | - James G McNamara
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Jane H Buckner
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, Washington, USA
| | - Srinath Sanda
- Immune Tolerance Network, Seattle, Washington, USA.,University of California, San Francisco, San Francisco, California, USA
| | | |
Collapse
|
24
|
Sun L, Yan Y, Lv H, Li J, Wang Z, Wang K, Wang L, Li Y, Jiang H, Zhang Y. Rapamycin targets STAT3 and impacts c-Myc to suppress tumor growth. Cell Chem Biol 2021; 29:373-385.e6. [PMID: 34706270 DOI: 10.1016/j.chembiol.2021.10.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/28/2021] [Accepted: 10/01/2021] [Indexed: 12/25/2022]
Abstract
Rapamycin is widely recognized as an inhibitor of mTOR, and has been approved for clinical use as an immunosuppressant. Its potencies in anti-cancer, anti-aging, and neurodegenerative diseases are emergingly established. The exploration of other targets of rapamycin will further elucidate its underlying mechanisms of action. In this study, we use a chemical proteomics strategy that has identified STAT3, a transcription factor considered to be undruggable, as a direct functional protein target of rapamycin. Together with other multi-dimensional proteomics data, we show that rapamycin treatment in cell culture significantly inhibits c-Myc-regulated gene expression. Furthermore, we show that rapamycin suppresses tumor growth along with a decreased expression of STAT3 and c-Myc in an in vivo xenograft mouse model for hepatocellular carcinoma. Our data suggest that rapamycin acts directly on STAT3 to decrease its transcription activity, providing important information for the pharmacological and pharmaceutical development of STAT3 inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Le Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Yan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Shanghai 201210, China
| | - Heng Lv
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianlong Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiyuan Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kun Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Wang
- ShanghaiTech University, Shanghai 201210, China
| | - Yunxia Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Shanghai 201210, China
| | - Hong Jiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Shanghai 201210, China
| | - Yaoyang Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Shanghai 201210, China.
| |
Collapse
|
25
|
Fu Y, Zhu F, Ma Z, Lv B, Wang X, Dai C, Ma X, Liu P, Lv H, Chen X, Chen Z, Shen L. Physalis alkekengi var. franchetii Extracts Exert Antitumor Effects on Non-Small Cell Lung Cancer and Multiple Myeloma by Inhibiting STAT3 Signaling. Onco Targets Ther 2021; 14:301-314. [PMID: 33469308 PMCID: PMC7811487 DOI: 10.2147/ott.s282334] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/29/2020] [Indexed: 12/04/2022] Open
Abstract
Background Physalis alkekengi var. franchetii is an herb that possesses various ethnopharmacological applications. Herein, our current study focuses on the antitumor effect of a combination of physalins, which are regarded as the most representative secondary metabolites from calyces of Physalis alkekengi var. franchetii. Materials and Methods We mainly investigated the antitumor activity of the physalins extracted from Physalis alkekengi var. franchetii on both solid and hematologic cancers. The main cells used in this study were NCI-H1975 and U266 cells. The major assays used were the CCK-8 assay, Western blot analyses, immunofluorescence assay and Annexin V assay, and a xenograft mouse model was used. Results The results showed that physalins exhibited a strong antitumoural effect on both non-small cell lung cancer (NSCLC) and multiple myeloma (MM) cells by suppressing constitutive STAT3 activity and further inhibiting the downstream target gene expression induced by STAT3 signaling, which resulted in the enhanced apoptosis of tumor cells. Moreover, physalins significantly reduced tumor growth in xenograft models of lung cancer. Conclusion Collectively, these findings demonstrated that the physalins from Physalis alkekengi var. franchetii may potentially act as cancer preventive or chemotherapeutic agents for NSCLC and MM by inhibiting the STAT3 signaling pathway. The present study served as a promising guide to further explore the precise mechanism of Physalis alkekengi var. franchetii in cancer treatment.
Collapse
Affiliation(s)
- Yufei Fu
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Insititute of Cancer Research, First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Fanfan Zhu
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Insititute of Cancer Research, First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Zhongjun Ma
- Institute of Marine Biology and Natural Products, Department of Ocean Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Bin Lv
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Insititute of Cancer Research, First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Xi Wang
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Insititute of Cancer Research, First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Chunyan Dai
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Insititute of Cancer Research, First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Xiaoqiong Ma
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Insititute of Cancer Research, First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Pei Liu
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Insititute of Cancer Research, First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Hang Lv
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Insititute of Cancer Research, First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Xin Chen
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China
| | - Zhe Chen
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Insititute of Cancer Research, First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Li Shen
- Institute of Basic Theory of Chinese Medicine, China Academy of Chinese Medicine Science, Beijing, People's Republic of China
| |
Collapse
|
26
|
Azar WS, Njeim R, Fares AH, Azar NS, Azar ST, El Sayed M, Eid AA. COVID-19 and diabetes mellitus: how one pandemic worsens the other. Rev Endocr Metab Disord 2020; 21:451-463. [PMID: 32743793 PMCID: PMC7395898 DOI: 10.1007/s11154-020-09573-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In light of the most challenging public health crisis of modern history, COVID-19 mortality continues to rise at an alarming rate. Patients with co-morbidities such as hypertension, cardiovascular disease, and diabetes mellitus (DM) seem to be more prone to severe symptoms and appear to have a higher mortality rate. In this review, we elucidate suggested mechanisms underlying the increased susceptibility of patients with diabetes to infection with SARS-CoV-2 with a more severe COVID-19 disease. The worsened prognosis of COVID-19 patients with DM can be attributed to a facilitated viral uptake assisted by the host's receptor angiotensin-converting enzyme 2 (ACE2). It can also be associated with a higher basal level of pro-inflammatory cytokines present in patients with diabetes, which enables a hyperinflammatory "cytokine storm" in response to the virus. This review also suggests a link between elevated levels of IL-6 and AMPK/mTOR signaling pathway and their role in exacerbating diabetes-induced complications and insulin resistance. If further studied, these findings could help identify novel therapeutic intervention strategies for patients with diabetes comorbid with COVID-19.
Collapse
Affiliation(s)
- William S Azar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut, 1107-2020, Lebanon
- AUB Diabetes, American University of Beirut, Beirut, Lebanon
- Department of Physiology and Biophysics, Georgetown University Medical Center, Washington, DC, USA
| | - Rachel Njeim
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut, 1107-2020, Lebanon
- AUB Diabetes, American University of Beirut, Beirut, Lebanon
| | - Angie H Fares
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut, 1107-2020, Lebanon
- AUB Diabetes, American University of Beirut, Beirut, Lebanon
| | - Nadim S Azar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut, 1107-2020, Lebanon
- AUB Diabetes, American University of Beirut, Beirut, Lebanon
| | - Sami T Azar
- AUB Diabetes, American University of Beirut, Beirut, Lebanon
- Department of Internal Medicine, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Mazen El Sayed
- Department of Emergency Medicine, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut, 1107-2020, Lebanon.
- AUB Diabetes, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
27
|
Feng B, Zhu Y, Yan L, Yan H, Huang X, Jiang D, Li Z, Hua L, Zhuo Y, Fang Z, Che L, Lin Y, Xu S, Huang C, Zou Y, Li L, Wu D. Ursolic acid induces the production of IL6 and chemokines in both adipocytes and adipose tissue. Adipocyte 2020; 9:523-534. [PMID: 32876525 PMCID: PMC7714451 DOI: 10.1080/21623945.2020.1814545] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/12/2020] [Accepted: 08/20/2020] [Indexed: 12/19/2022] Open
Abstract
Adipose tissue inflammation plays an important role in the regulation of glucose and lipids metabolism. It is unknown whether Ursolic acid (UA) could regulate adipose tissue inflammation, though it can regulate inflammation in many other tissues. In this study, 3T3-L1 adipocytes, DIO mice and lean mice were treated with UA or vehicle. Gene expression of inflammatory factors, chemokines and immune markers in adipocytes and adipose tissue, cytokines in cell culture medium and serum, and inflammation regulatory pathways in adipocytes were detected. Results showed that UA increased the expression of interleukins and chemokines, but not TNFα, in both adipocytes and adipose tissue. IL6 and MCP1 levels in the cell culture medium and mouse serum were induced by UA treatment. Cd14 expression level and number of CD14+ monocytes were higher in UA treated adipose tissue than those in the control group. Glucose tolerance test was impaired by UA treatment in DIO mice. Mechanistically, UA induced the expression of Tlr4 and the phosphorylation levels of ERK and NFκB in adipocytes. In conclusion, our study indicated that short-term UA administration could induce CD14+ monocytes infiltration by increasing the production of interleukins and chemokines in mouse adipose tissue, which might further impair glucose tolerance test.
Collapse
Affiliation(s)
- Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yingguo Zhu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lijun Yan
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hui Yan
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaohua Huang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dandan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhen Li
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lun Hua
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Chao Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yuanfeng Zou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lixia Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
28
|
He Y, Fan Z, He L, Zhang C, Ping F, Deng M, Liu S, Wang Y, Cheng B, Xia J. Metformin Combined with 4SC-202 Inhibited the Migration and Invasion of OSCC via STAT3/TWIST1. Onco Targets Ther 2020; 13:11019-11029. [PMID: 33149616 PMCID: PMC7605634 DOI: 10.2147/ott.s268851] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/11/2020] [Indexed: 01/27/2023] Open
Abstract
Background Oral squamous cell carcinoma (OSCC), the most common epithelial malignant neoplasm in the head and neck, characterizes with local infiltration and metastasis of lymph nodes. The five-year survival rate of OSCC remains low despite the advances in clinical methods. Thus, it is necessary to develop a new effective therapeutic scheme for OSCC. Our previous results showed that metformin and 4SC-202 synergistically promoted the intrinsic apoptosis of OSCC in vitro and in vivo, but the effects on invasion and migration remained unclear. Methods Human OSCC cell lines HSC6 and CAL33 were cultured with metformin (16 mM) or/and 4SC-202 (0.4 μM) for 72 h. STAT3 inhibitor S31-201 was applied at concentration of 60 μM for 48 h. Wound-healing assays and transwell assays were used to determine the invasion and migration ability of OSCC. qRT-PCR and Western blot were performed to detect mRNA levels and protein levels. Results Metformin or/and 4SC-202 suppressed the migration and invasion of OSCC cells. Importantly, the expression of TWIST1 was suppressed by metformin and 4SC-202, while the invasion and migration inhibitory effects of metformin and 4SC-202 were countered by the overexpression of TWIST1. In addition, the phosphorylation level of STAT3 decreased after the administration of metformin or/and 4SC-202. Furthermore, inhibition of STAT3 by S31-201 suppressed the expression of TWIST1 and led to a decline in migration and invasion of OSCC, while overexpression of TWIST1 attenuated these effects. Conclusion Metformin and 4SC-202 suppressed the invasion and migration of OSCC through inhibition of STAT3/TWIST1, and this scheme can serve as a novel therapeutic strategy for OSCC.
Collapse
Affiliation(s)
- Yuan He
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zhaona Fan
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Lihong He
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Chi Zhang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Fan Ping
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Miao Deng
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Suyang Liu
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Yanting Wang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| |
Collapse
|
29
|
Liao KY, Chen CJ, Hsieh SK, Pan PH, Chen WY. Interleukin-13 ameliorates postischemic hepatic gluconeogenesis and hyperglycemia in rat model of stroke. Metab Brain Dis 2020; 35:1201-1210. [PMID: 32632665 DOI: 10.1007/s11011-020-00596-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022]
Abstract
Hyperglycemia is a well-known indicator of stroke prognosis, and one-third of nondiabetic patients develop postischemic hyperglycemia during the acute phase of stroke; this is related to relatively poor prognosis, high mortality, and impaired neurological recovery. Interleukin-13 (IL-13), a member of the Th2 cytokine family, is involved in both the regulation of immune response and glucose metabolism. Thus, we investigated the mechanism of postischemic hyperglycemia and the role of IL-13 by using a permanent middle cerebral artery occlusion (MCAO) rat model. Our results indicated that postischemic hyperglycemia was accompanied with hyperinsulinemia and increased HOMA-IR, elevated hepatic gluconeogenesis, and suppressed insulin signaling. A shift towards inflammatory response was evident with results of elevated proinflammatory cytokines and increased expression of negative regulatory proteins, suggesting an ongoing vicious cycle of inflammatory-induced insulin-resistant hyperglycemia. IL-13 treatment counteracted the proinflammatory states and abolished the vicious cycle through enhancing STAT6 and STAT3, which mediated the immune and metabolic pathways respectively; these effects resolved the formerly described pathological changes of postischemic hyperglycemia and reduced infarction size in the MCAO rats. Our findings demonstrated the importance of Th1-Th2 balance in the peripheral glucose metabolism affected by acute ischemic stroke, which provides a new perspective for the prevention and control of postischemic hyperglycemia.
Collapse
Affiliation(s)
- Keng-Ying Liao
- Department of Veterinary Medicine, National Chung Hsing University, 145 Xingda Rd., Taichung City, South Dist., 402, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Sheng-Kuo Hsieh
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Ping-Ho Pan
- Department of Veterinary Medicine, National Chung Hsing University, 145 Xingda Rd., Taichung City, South Dist., 402, Taiwan
- Department of Pediatrics, Tungs' Taichung Metro Harbor Hospital, Taichung, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, 145 Xingda Rd., Taichung City, South Dist., 402, Taiwan.
| |
Collapse
|
30
|
Indoximod opposes the immunosuppressive effects mediated by IDO and TDO via modulation of AhR function and activation of mTORC1. Oncotarget 2020; 11:2438-2461. [PMID: 32637034 PMCID: PMC7321702 DOI: 10.18632/oncotarget.27646] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022] Open
Abstract
Indoximod has shaped our understanding of the biology of IDO1 in the control of immune responses, though its mechanism of action has been poorly understood. Previous studies demonstrated that indoximod creates a tryptophan (Trp) sufficiency signal that reactivates mTOR in the context of low Trp concentrations, thus opposing the effects caused by IDO1. Here we extend the understanding of indoximod’s mechanism of action by showing that it has pleiotropic effects on immune regulation. Indoximod can have a direct effect on T cells, increasing their proliferation as a result of mTOR reactivation. Further, indoximod modulates the differentiation of CD4+ T cells via the aryl hydrocarbon receptor (AhR), which controls transcription of several genes in response to different ligands including kynurenine (Kyn). Indoximod increases the transcription of RORC while inhibiting transcription of FOXP3, thus favoring differentiation to IL-17-producing helper T cells and inhibiting the differentiation of regulatory T cells. These indoximod-driven effects on CD8+ and CD4+ T cells were independent from the activity of IDO/TDO and from the presence of exogenous Kyn, though they do oppose the effects of Kyn produced by these Trp catabolizing enzymes. Indoximod can also downregulate expression of IDO protein in vivo in murine lymph node dendritic cells and in vitro in human monocyte-derived dendritic cells via a mechanism that involves signaling through the AhR. Together, these data improve the understanding of how indoximod influences the effects of IDO, beyond and distinct from direct enzymatic inhibition of the enzyme.
Collapse
|
31
|
Cai WJ, Liang XF, Yuan XC, Li AX, He S. Changes of DNA Methylation Pattern in Metabolic Pathways Induced by High-Carbohydrate Diet Contribute to Hyperglycemia and Fat Deposition in Grass Carp ( Ctenopharyngodon idellus). Front Endocrinol (Lausanne) 2020; 11:398. [PMID: 32754117 PMCID: PMC7381294 DOI: 10.3389/fendo.2020.00398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 05/18/2020] [Indexed: 12/21/2022] Open
Abstract
Although studies have determined that epigenetics plays an essential role in regulating metabolism in mammals, research on nutrition-related DNA methylation remains to be lacking in teleosts. In the present study, we provided a hepatic whole-genome DNA methylation analysis in grass carp fed with moderate- or excessive-carbohydrate-level diet. Although a high-carbohydrate (HC) diet significantly changed the mRNA expression levels of metabolic genes, it did not affect the global genomic DNA methylation levels in grass carp liver. However, compared with the control group, 3,972 genes were hyper-methylated and 2,904 genes were hypo-methylated in the promoter region. Meanwhile, 10,711 genes were hyper-methylated and 6,764 genes were hypo-methylated in the gene body region in the HC group. These differentially methylated genes (DMGs) were enriched in multiple pathways, including carbohydrate metabolism, insulin pathway, lipid metabolism, and adipocytokine signaling pathway. In addition, the variations in DNA methylation significantly regulated the transcription levels of key genes of metabolism, which could affect the glucose concentrations and the lipid deposition of grass carp. Furthermore, we compared the DNA methylation alterations of genes in glucose metabolism and obesity pathways of grass carp with those of mammalian models in different nutritional states. The results showed that most of the DMGs in grass carp were also regulated by DNA methylation in mammals when the nutritional state changed. The findings revealed more differentially methylated regions and candidate genes for glucose metabolism and broken species boundaries.
Collapse
Affiliation(s)
- Wen-Jing Cai
- Chinese Perch Research Center, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, China
| | - Xu-Fang Liang
- Chinese Perch Research Center, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, China
- *Correspondence: Xu-Fang Liang
| | - Xiao-Chen Yuan
- Chinese Perch Research Center, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, China
| | - Ai-Xuan Li
- Chinese Perch Research Center, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, China
| | - Shan He
- Chinese Perch Research Center, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, China
| |
Collapse
|
32
|
Syring KE, Cyphert TJ, Beck TC, Flynn CR, Mignemi NA, McGuinness OP. Systemic bile acids induce insulin resistance in a TGR5-independent manner. Am J Physiol Endocrinol Metab 2019; 316:E782-E793. [PMID: 30779633 PMCID: PMC6732652 DOI: 10.1152/ajpendo.00362.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 02/04/2019] [Accepted: 02/18/2019] [Indexed: 02/07/2023]
Abstract
Bile acids are involved in the emulsification and absorption of dietary fats, as well as acting as signaling molecules. Recently, bile acid signaling through farnesoid X receptor and G protein-coupled bile acid receptor (TGR5) has been reported to elicit changes in not only bile acid synthesis but also metabolic processes, including the alteration of gluconeogenic gene expression and energy expenditure. A role for bile acids in glucose metabolism is also supported by a correlation between changes in the metabolic state of patients (i.e., obesity or postbariatric surgery) and altered serum bile acid levels. However, despite evidence for a role for bile acids during metabolically challenging settings, the direct effect of elevated bile acids on insulin action in the absence of metabolic disease has yet to be investigated. The present study examines the impact of acutely elevated plasma bile acid levels on insulin sensitivity using hyperinsulinemic-euglycemic clamps. In wild-type mice, elevated bile acids impair hepatic insulin sensitivity by blunting the insulin suppression of hepatic glucose production. The impaired hepatic insulin sensitivity could not be attributed to TGR5 signaling, as TGR5 knockout mice exhibited a similar inhibition of insulin suppression of hepatic glucose production. Canonical insulin signaling pathways, such as hepatic PKB (or Akt) activation, were not perturbed in these animals. Interestingly, bile acid infusion directly into the portal vein did not result in an impairment in hepatic insulin sensitivity. Overall, the data indicate that acute increases in circulating bile acids in lean mice impair hepatic insulin sensitivity via an indirect mechanism.
Collapse
Affiliation(s)
- Kristen E Syring
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Travis J Cyphert
- Department of Biological Sciences, Marshall University College of Science, Huntington, West Virginia
| | - Thomas C Beck
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Charles R Flynn
- Department of Surgery, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Nicholas A Mignemi
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine , Nashville, Tennessee
| |
Collapse
|
33
|
Heo YJ, Choi SE, Jeon JY, Han SJ, Kim DJ, Kang Y, Lee KW, Kim HJ. Visfatin Induces Inflammation and Insulin Resistance via the NF- κB and STAT3 Signaling Pathways in Hepatocytes. J Diabetes Res 2019; 2019:4021623. [PMID: 31396538 PMCID: PMC6664505 DOI: 10.1155/2019/4021623] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/09/2019] [Accepted: 06/10/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND It has been suggested that visfatin, which is an adipocytokine, exhibits proinflammatory properties and is associated with insulin resistance. Insulin resistance and inflammation are the principal pathogeneses of nonalcoholic fatty liver disease (NAFLD), but the relationship, if any, between visfatin and NAFLD remains unclear. Here, we evaluated the effects of visfatin on hepatic inflammation and insulin resistance in HepG2 cells and examined the molecular mechanisms involved. METHODS After treatment with visfatin, the inflammatory cytokines IL-6, TNF-α, and IL-1β were assessed by real-time polymerase chain reaction (RT-PCR) and immunocytochemical staining in HepG2 cells. To investigate the effects of visfatin on insulin resistance, we evaluated insulin-signaling pathways, such as IR, IRS-1, GSK, and AKT using immunoblotting. We assessed the intracellular signaling molecules including STAT3, NF-κB, IKK, p38, JNK, and ERK by western blotting. We treated HepG2 cells with both visfatin and either AG490 (a JAK2 inhibitor) or Bay 7082 (an NF-κB inhibitor); we examined proinflammatory cytokine mRNA levels using RT-PCR and insulin signaling using western blotting. RESULTS In HepG2 cells, visfatin significantly increased the levels of proinflammatory cytokines, reduced the levels of proteins (e.g., phospho-IR, phospho-IRS-1 (Tyr612), phospho-AKT, and phospho-GSK-3α/β) involved in insulin signaling, and increased IRS-1 S307 phosphorylation compared to controls. Interestingly, visfatin increased the activities of the JAK2/STAT3 and IKK/NF-κB signaling pathways but not those of the JNK, p38, and ERK pathways. Visfatin-induced inflammation and insulin resistance were regulated by JAK2/STAT3 and IKK/NF-κB signaling; together with AG490 or Bay 7082, visfatin significantly reduced mRNA levels of IL-6, TNF-α and IL-1β and rescued insulin signaling. CONCLUSION Visfatin induced proinflammatory cytokine production and inhibited insulin signaling via the STAT3 and NF-κB pathways in HepG2 cells.
Collapse
Affiliation(s)
- Yu Jung Heo
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Sung-E Choi
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Ja Young Jeon
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Dae Jung Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yup Kang
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Kwan Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hae Jin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
34
|
Zegeye MM, Lindkvist M, Fälker K, Kumawat AK, Paramel G, Grenegård M, Sirsjö A, Ljungberg LU. Activation of the JAK/STAT3 and PI3K/AKT pathways are crucial for IL-6 trans-signaling-mediated pro-inflammatory response in human vascular endothelial cells. Cell Commun Signal 2018; 16:55. [PMID: 30185178 PMCID: PMC6125866 DOI: 10.1186/s12964-018-0268-4] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/27/2018] [Indexed: 01/05/2023] Open
Abstract
Background IL-6 classic signaling is linked to anti-inflammatory functions while the trans-signaling is associated with pro-inflammatory responses. Classic signaling is induced via membrane-bound IL-6 receptor (IL-6R) whereas trans-signaling requires prior binding of IL-6 to the soluble IL-6R. In both cases, association with the signal transducing gp130 receptor is compulsory. However, differences in the downstream signaling mechanisms of IL-6 classic- versus trans-signaling remains largely elusive. Methods In this study, we used flow cytometry, quantitative PCR, ELISA and immuno-blotting techniques to investigate IL-6 classic and trans-signaling mechanisms in Human Umbilical Vein Endothelial Cells (HUVECs). Results We show that both IL-6R and gp130 are expressed on the surface of human vascular endothelial cells, and that the expression is affected by pro-inflammatory stimuli. In contrast to IL-6 classic signaling, IL-6 trans-signaling induces the release of the pro-inflammatory chemokine Monocyte Chemoattractant Protein-1 (MCP-1) from human vascular endothelial cells. In addition, we reveal that the classic signaling induces activation of the JAK/STAT3 pathway while trans-signaling also activates the PI3K/AKT and the MEK/ERK pathways. Furthermore, we demonstrate that MCP-1 induction by IL-6 trans-signaling requires simultaneous activation of the JAK/STAT3 and PI3K/AKT pathways. Conclusions Collectively, our study reports molecular differences in IL-6 classic- and trans-signaling in human vascular endothelial cells; and elucidates the pathways which mediate MCP-1 induction by IL-6 trans-signaling. Electronic supplementary material The online version of this article (10.1186/s12964-018-0268-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mulugeta M Zegeye
- Cardiovascular Research Center, School of Medical Sciences, Örebro University Södra Grev Rosengatan 32, 703 62, Örebro, Sweden.
| | - Madelene Lindkvist
- Cardiovascular Research Center, School of Medical Sciences, Örebro University Södra Grev Rosengatan 32, 703 62, Örebro, Sweden
| | - Knut Fälker
- Cardiovascular Research Center, School of Medical Sciences, Örebro University Södra Grev Rosengatan 32, 703 62, Örebro, Sweden
| | - Ashok K Kumawat
- Cardiovascular Research Center, School of Medical Sciences, Örebro University Södra Grev Rosengatan 32, 703 62, Örebro, Sweden
| | - Geena Paramel
- Cardiovascular Research Center, School of Medical Sciences, Örebro University Södra Grev Rosengatan 32, 703 62, Örebro, Sweden.,Present address: Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, E2L 4L5, Canada
| | - Magnus Grenegård
- Cardiovascular Research Center, School of Medical Sciences, Örebro University Södra Grev Rosengatan 32, 703 62, Örebro, Sweden
| | - Allan Sirsjö
- Cardiovascular Research Center, School of Medical Sciences, Örebro University Södra Grev Rosengatan 32, 703 62, Örebro, Sweden
| | - Liza U Ljungberg
- Cardiovascular Research Center, School of Medical Sciences, Örebro University Södra Grev Rosengatan 32, 703 62, Örebro, Sweden
| |
Collapse
|
35
|
Chronic stress of high dietary carbohydrate level causes inflammation and influences glucose transport through SOCS3 in Japanese flounder Paralichthys olivaceus. Sci Rep 2018; 8:7415. [PMID: 29743495 PMCID: PMC5943576 DOI: 10.1038/s41598-018-25412-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/13/2018] [Indexed: 02/06/2023] Open
Abstract
Carnivorous fish is thought to be high-glucose intolerance. But the reasons were still unclear. The aim of the present study is to investigate the effects of high level of dietary carbohydrate on the survival, growth and immune responses of Paralichthys olivaceus, and the underlying molecular mechanism related to the immune and glucose metabolism. P. olivaceus were fed with 8%, 16% and 24% of dietary carbohydrate for 10 weeks, respectively. After that, a glucose tolerance test (GTT) was conducted. Results showed that excessive (24%) dietary carbohydrate significantly decreased the growth and glucose tolerance ability according to the GTT. It significantly increased hepatic NADPH oxidase activity and malondialdehyde content and serum contents of IL-6 and advanced glycation end products. The expressions of glucose transport-relevant genes in liver and the content of related hormones in serum were analyzed. In conclusion, it was confirmed that IL-6 increased the expression of suppressor of cytokine signaling 3 (SOCS3) and regulated the downstream targets of PI3K-AKT mediated signal transduction, and then downregulated the glucose transporter 2 activity in liver of P. olivaceus fed diet with excessive carbohydrate level. It was suggested that SOCS3 served as a bridge between immune response and glucose metabolism in P. olivaceus.
Collapse
|
36
|
Makwana K, Patel SA, Velingkaar N, Ebron JS, Shukla GC, Kondratov RVKV. Aging and calorie restriction regulate the expression of miR-125a-5p and its target genes Stat3, Casp2 and Stard13. Aging (Albany NY) 2018; 9:1825-1843. [PMID: 28783714 PMCID: PMC5559175 DOI: 10.18632/aging.101270] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 07/27/2017] [Indexed: 12/18/2022]
Abstract
Calorie restriction (CR) is a dietary intervention known to delay aging. In order, to understand molecular mechanisms of CR, we analyzed the expression of 983 MicroRNAs (miRNAs) in the liver of female mice after 2 years of 30% CR using micro-array. 16 miRNAs demonstrated significant changes in their expression upon CR in comparison with age-matched control. mmu-miR-125a-5p (miR-125a-5p) was significantly upregulated upon CR, and in agreement with this, the expression of mRNAs for its three predicted target genes: Stat3, Casp2, and Stard13 was significantly downregulated in the liver of CR animals. The expression of precursor miRNA for miR-125a-5p was also upregulated upon CR, which suggests its regulation at the level of transcription. Upon aging miR-125a-5p expression was downregulated while the expression of its target genes was upregulated. Thus, CR prevented age-associated changes in the expression of miR-125a-5p and its targets. We propose that miR-125a-5p dependent downregulation of Stat3, Casp2, and Stard13 contributes to the calorie restriction-mediated delay of aging.
Collapse
Affiliation(s)
- Kuldeep Makwana
- Center for Gene Regulation in Health and Disease and Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Sonal Arvind Patel
- Center for Gene Regulation in Health and Disease and Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Nikkhil Velingkaar
- Center for Gene Regulation in Health and Disease and Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Jey Sabith Ebron
- Center for Gene Regulation in Health and Disease and Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Girish C Shukla
- Center for Gene Regulation in Health and Disease and Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Roman V Kondratov V Kondratov
- Center for Gene Regulation in Health and Disease and Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| |
Collapse
|
37
|
IL-6 signalling pathways and the development of type 2 diabetes. Inflammopharmacology 2018; 26:685-698. [PMID: 29508109 DOI: 10.1007/s10787-018-0458-0] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/21/2018] [Indexed: 02/07/2023]
Abstract
Interleukin 6 (IL-6), a multifunctional cytokine, has been implicated in the pathophysiology of type 2 diabetes (T2D). The elevated circulating level of IL-6 is an independent predictor of T2D and is considered to be involved in the development of inflammation, insulin resistance and β-cell dysfunction. On the other hand, an increasing number of evidence suggests that IL-6 has an anti-inflammatory role and improves glucose metabolism. The complex signal transduction mechanism of IL-6 may help explain the pleiotropic nature of the cytokine. IL-6 acts via two distinct signalling pathways called classic signalling and trans-signalling. While both signalling modes lead to activation of the same receptor subunit, their final biological effects are completely different. The aim of this review is to summarize our current knowledge about the role of IL-6 in the development of T2D. We will also discuss the importance of specific blockade of IL-6 trans-signalling rather than inhibiting both signalling pathways as a therapeutic strategy for the treatment of T2D and its associated macrovascular complications.
Collapse
|
38
|
Yoo T, Ham SA, Lee WJ, Hwang SI, Park JA, Hwang JS, Hur J, Shin HC, Han SG, Lee CH, Han DW, Paek KS, Seo HG. Ligand-Dependent Interaction of PPARδ With T-Cell Protein Tyrosine Phosphatase 45 Enhances Insulin Signaling. Diabetes 2018; 67:360-371. [PMID: 29233935 DOI: 10.2337/db17-0499] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 11/27/2017] [Indexed: 12/16/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR) δ plays a pivotal role in metabolic homeostasis through its effect on insulin signaling. Although diverse genomic actions of PPARδ are postulated, the specific molecular mechanisms whereby PPARδ controls insulin signaling have not been fully elucidated. We demonstrate here that short-term activation of PPARδ results in the formation of a stable complex with nuclear T-cell protein tyrosine phosphatase 45 (TCPTP45) isoform. This interaction of PPARδ with TCPTP45 blocked translocation of TCPTP45 into the cytoplasm, thereby preventing its interaction with the insulin receptor, which inhibits insulin signaling. Interaction of PPARδ with TCPTP45 blunted interleukin 6-induced insulin resistance, leading to retention of TCPTP45 in the nucleus, thereby facilitating deactivation of the signal transducer and activator of transcription 3 (STAT3)-suppressor of cytokine signaling 3 (SOCS3) signal. Finally, GW501516-activated PPARδ improved insulin signaling and glucose intolerance in mice fed a high-fat diet through its interaction with TCPTP45. This novel interaction of PPARδ constitutes the most upstream component identified of the mechanism downregulating insulin signaling.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Adipocytes, White/drug effects
- Adipocytes, White/immunology
- Adipocytes, White/metabolism
- Adipocytes, White/pathology
- Alternative Splicing
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Cell Line
- Cells, Cultured
- Glucose Intolerance/etiology
- Glucose Intolerance/immunology
- Glucose Intolerance/prevention & control
- Hepatocytes/drug effects
- Hepatocytes/immunology
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Humans
- Insulin Resistance
- Male
- Mice, Inbred ICR
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/immunology
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Obesity/drug therapy
- Obesity/metabolism
- Obesity/pathology
- Obesity/physiopathology
- PPAR delta/agonists
- PPAR delta/antagonists & inhibitors
- PPAR delta/genetics
- PPAR delta/metabolism
- Protein Multimerization/drug effects
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/chemistry
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism
- RNA Interference
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/metabolism
- Specific Pathogen-Free Organisms
- Thiazoles/pharmacology
- Thiazoles/therapeutic use
Collapse
Affiliation(s)
- Taesik Yoo
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Sun Ah Ham
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Won Jin Lee
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Seon In Hwang
- Department of Stem Cell Biology, Konkuk University, Seoul, Korea
| | - Jin-A Park
- Department of Veterinary Pharmacology and Toxicology, Konkuk University, Seoul, Korea
| | - Jung Seok Hwang
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Jinwoo Hur
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Ho-Chul Shin
- Department of Veterinary Pharmacology and Toxicology, Konkuk University, Seoul, Korea
| | - Sung Gu Han
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Chi-Ho Lee
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Dong Wook Han
- Department of Stem Cell Biology, Konkuk University, Seoul, Korea
| | | | - Han Geuk Seo
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| |
Collapse
|
39
|
Giguère V. Canonical signaling and nuclear activity of mTOR-a teamwork effort to regulate metabolism and cell growth. FEBS J 2018; 285:1572-1588. [PMID: 29337437 DOI: 10.1111/febs.14384] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/21/2017] [Accepted: 01/10/2018] [Indexed: 01/07/2023]
Abstract
Mechanistic (or mammalian) target of rapamycin (mTOR) is a kinase that regulates almost all functions related to cell growth and metabolism in response to extra- and intracellular stimuli, such as availability of nutrients, the presence of growth factors, or the energy status of the cell. As part of two distinct protein complexes, mTORC1 and mTORC2, the kinase has been shown to influence cell growth and proliferation by controlling ribosome biogenesis, mRNA translation, carbohydrate and lipid metabolism, protein degradation, autophagy as well as microtubule and actin dynamics. In addition to these well-characterized functions, mTOR can also influence gene transcription. While most studies focused on investigating how canonical mTOR signaling regulates the activity of transcription factors outside the nucleus, recent findings point to a more direct role for mTOR as a transcription factor operating on chromatin in the nucleus. In particular, recent genome-wide identification of mTOR targets on chromatin reveals that its activities in the nucleus and cytoplasm are functionally and biologically linked, thus uncovering a novel paradigm in mTOR function.
Collapse
Affiliation(s)
- Vincent Giguère
- Departments of Biochemistry, Medicine and Oncology, Faculty of Medicine, Goodman Cancer Research Centre, McGill University, Montréal, Canada
| |
Collapse
|
40
|
Chen L, Tang Z, Wang X, Ma H, Shan D, Cui S. PKM2 aggravates palmitate-induced insulin resistance in HepG2 cells via STAT3 pathway. Biochem Biophys Res Commun 2017; 492:109-115. [PMID: 28802581 DOI: 10.1016/j.bbrc.2017.08.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 08/08/2017] [Indexed: 01/01/2023]
Abstract
Studies have identified that PKM2 is related to the development of glucose intolerance and insulin resistance in rodents and humans. However, the underlying mechanism remains largely unknown. In the present study, we found that PKM2 expression was significantly elevated in insulin-resistant hepatic tissues and hepatocytes, implicating an association between PKM2 expression and hepatic insulin resistance (IR). In vitro study revealed that overexpression of PKM2 impaired the insulin signaling pathway by decreasing the phosphorylation of protein kinase B (Akt) and glycogen synthase kinase-3β (GSK3β). Furthermore, PKM2 overexpression enhanced the effects of PA on the lipid accumulation, the expression of phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase) and hepatic glucose uptake. Intriguingly, PA-induced insulin resistance was suppressed following by the ablation of PKM2 in HepG2 cells. We also found that STAT3 was significantly activated by PKM2 overexpression. Moreover, we identified that PKM2 could interact directly with STAT3. Taken together, these studies demonstrate that PKM2 may promote hepatic IR via STAT3 pathway and would provide a new insight into dissecting the molecular pathogenesis of hepatic insulin resistance.
Collapse
Affiliation(s)
- Ling Chen
- Department of Endocrinology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu Province, People's Republic of China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Zhuqi Tang
- Department of Endocrinology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Xiaohua Wang
- Department of Endocrinology, The Second Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Hong Ma
- Department of Endocrinology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu Province, People's Republic of China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Dandan Shan
- Department of Endocrinology, The Second Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Shiwei Cui
- Department of Endocrinology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu Province, People's Republic of China.
| |
Collapse
|
41
|
Dutheil F, Gordon BA, Naughton G, Crendal E, Courteix D, Chaplais E, Thivel D, Lac G, Benson AC. Cardiovascular risk of adipokines: a review. J Int Med Res 2017; 46:2082-2095. [PMID: 28974138 PMCID: PMC6023062 DOI: 10.1177/0300060517706578] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Over the last two decades, the understanding of adipose tissue has undergone radical change. The perception has evolved from an inert energy storage tissue to that of an active endocrine organ. Adipose tissue releases a cluster of active molecules named adipokines. The severity of obesity-related diseases does not necessarily correlate with the extent of body fat accumulation but is closely related to body fat distribution, particularly to visceral localization. There is a distinction between the metabolic function of central obesity (visceral abdominal) and peripheral obesity (subcutaneous) in the production of adipokines. Visceral fat accumulation, linked with levels of some adipokines, induces chronic inflammation and metabolic disorders, including glucose intolerance, hyperlipidaemia, and arterial hypertension. Together, these conditions contribute to a diagnosis of metabolic syndrome, directly associated with the onset of cardiovascular disease. If it is well known that adipokines contribute to the inflammatory profile and appetite regulation, this review is novel in synthesising the current state of knowledge of the role of visceral adipose tissue and its secretion of adipokines in cardiovascular risk.
Collapse
Affiliation(s)
- Frédéric Dutheil
- 1 University Hospital of Clermont-Ferrand (CHU), Preventive and Occupational Medicine, Clermont-Ferrand, France.,2 Université Clermont Auvergne, CNRS, LaPSCo, Physiological and Psychosocial Stress, Clermont-Ferrand, France.,3 Australian Catholic University, Faculty of Health, School of Exercise Science, Melbourne, Victoria, Australia.,4 WittyFit, Paris, France
| | - Brett Ashley Gordon
- 5 La Trobe University, La Trobe Rural Health School, Exercise Physiology, Bendigo, Victoria, Australia
| | - Geraldine Naughton
- 3 Australian Catholic University, Faculty of Health, School of Exercise Science, Melbourne, Victoria, Australia
| | - Edward Crendal
- 3 Australian Catholic University, Faculty of Health, School of Exercise Science, Melbourne, Victoria, Australia
| | - Daniel Courteix
- 3 Australian Catholic University, Faculty of Health, School of Exercise Science, Melbourne, Victoria, Australia.,6 Université Clermont Auvergne, Laboratoire des Adaptations Métaboliques à l'Exercice en conditions Physiologiques et Pathologiques (AME2P), Clermont-Ferrand, France
| | - Elodie Chaplais
- 3 Australian Catholic University, Faculty of Health, School of Exercise Science, Melbourne, Victoria, Australia.,6 Université Clermont Auvergne, Laboratoire des Adaptations Métaboliques à l'Exercice en conditions Physiologiques et Pathologiques (AME2P), Clermont-Ferrand, France
| | - David Thivel
- 6 Université Clermont Auvergne, Laboratoire des Adaptations Métaboliques à l'Exercice en conditions Physiologiques et Pathologiques (AME2P), Clermont-Ferrand, France
| | - Gérard Lac
- 6 Université Clermont Auvergne, Laboratoire des Adaptations Métaboliques à l'Exercice en conditions Physiologiques et Pathologiques (AME2P), Clermont-Ferrand, France
| | | |
Collapse
|
42
|
Shan M, Qin J, Jin F, Han X, Guan H, Li X, Zhang J, Zhang H, Wang Y. Autophagy suppresses isoprenaline-induced M2 macrophage polarization via the ROS/ERK and mTOR signaling pathway. Free Radic Biol Med 2017. [PMID: 28647611 DOI: 10.1016/j.freeradbiomed.2017.05.021] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The objective of this study was to examine the effect of autophagy on stress-induced M2 macrophage polarization in the tumor microenvironment of breast cancer and to determine whether the underlying mechanism was related to the reactive oxygen species (ROS)/ERK and mTOR pathway. In vitro, we found that the basal autophagy level in mouse RAW 264.7 macrophages decreased with the incubation of tumor cell culture supernatant. Similarly, the polarization of RAW 264.7 to M2 macrophages was inhibited by the autophagy inducer rapamycin and increased by the autophagy inhibitor 3-MA or by siBeclin1. In addition, we found that not only was M2 molecule expression down-regulated but intracellular ROS generation was also blocked by autophagy induction. In vivo, we observed that mice that received an isoprenaline injection as a stress agent exhibited augmented implanted breast tumor growth, lung metastasis, intratumoral mRNA expression of M2 molecules and serum ROS generation. In contrast, the intratumoral expression of LC3-II and Beclin1 was decreased. In addition, we observed that isoprenaline induced the up-regulation of the intratumoral expression of phosphorylated mTOR, phosphorylated ERK1/2, phosphorylated Tyr705-STAT3 and HIF-1α, whereas rapamycin induced an opposite effect on the same molecules and could abolish the effects of isoprenaline. These results suggest that autophagy might suppress M2 macrophage polarization induced by isoprenaline via the ROS/ERK and mTOR signaling pathway. Our findings provide a theoretical basis for why high levels of stress hormones accelerate the progression of breast cancer, and autophagy may play a role in determining the outcomes of cancer therapy.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- Adenine/pharmacology
- Animals
- Antibiotics, Antineoplastic/pharmacology
- Autophagy/drug effects
- Autophagy/genetics
- Cell Line, Tumor
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Isoproterenol/antagonists & inhibitors
- Isoproterenol/pharmacology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- MAP Kinase Signaling System
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Microtubule-Associated Proteins/genetics
- Microtubule-Associated Proteins/metabolism
- RAW 264.7 Cells
- Reactive Oxygen Species/antagonists & inhibitors
- Reactive Oxygen Species/metabolism
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Sirolimus/pharmacology
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Tumor Microenvironment/drug effects
Collapse
Affiliation(s)
- Meihua Shan
- Medical School of Nankai University, Nankai University, Tianjin, China; Hebei Normal University for Nationalities, Chengde, China
| | - Junfang Qin
- Medical School of Nankai University, Nankai University, Tianjin, China
| | - Fengjiao Jin
- Medical School of Nankai University, Nankai University, Tianjin, China
| | - Xiao Han
- Medical School of Nankai University, Nankai University, Tianjin, China
| | - Haitao Guan
- Medical School of Nankai University, Nankai University, Tianjin, China
| | - Xiaoge Li
- Medical School of Nankai University, Nankai University, Tianjin, China
| | - Jiahui Zhang
- Medical School of Nankai University, Nankai University, Tianjin, China
| | - Hongyao Zhang
- Medical School of Nankai University, Nankai University, Tianjin, China
| | - Yue Wang
- Medical School of Nankai University, Nankai University, Tianjin, China.
| |
Collapse
|
43
|
Gao S, Durstine JL, Koh HJ, Carver WE, Frizzell N, Carson JA. Acute myotube protein synthesis regulation by IL-6-related cytokines. Am J Physiol Cell Physiol 2017; 313:C487-C500. [PMID: 28768641 DOI: 10.1152/ajpcell.00112.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/20/2017] [Accepted: 07/25/2017] [Indexed: 12/19/2022]
Abstract
IL-6 and leukemia inhibitory factor (LIF), members of the IL-6 family of cytokines, play recognized paradoxical roles in skeletal muscle mass regulation, being associated with both growth and atrophy. Overload or muscle contractions can induce a transient increase in muscle IL-6 and LIF expression, which has a regulatory role in muscle hypertrophy. However, the cellular mechanisms involved in this regulation have not been completely identified. The induction of mammalian target of rapamycin complex 1 (mTORC1)-dependent myofiber protein synthesis is an established regulator of muscle hypertrophy, but the involvement of the IL-6 family of cytokines in this process is poorly understood. Therefore, we investigated the acute effects of IL-6 and LIF administration on mTORC1 signaling and protein synthesis in C2C12 myotubes. The role of glycoprotein 130 (gp130) receptor and downstream signaling pathways, including phosphoinositide 3-kinase (PI3K)-Akt-mTORC1 and signal transducer and activator of transcription 3 (STAT3)-suppressor of cytokine signaling 3 (SOCS3), was investigated by administration of specific siRNA or pharmaceutical inhibitors. Acute administration of IL-6 and LIF induced protein synthesis, which was accompanied by STAT3 activation, Akt-mTORC1 activation, and increased SOCS3 expression. This induction of protein synthesis was blocked by both gp130 siRNA knockdown and Akt inhibition. Interestingly, STAT3 inhibition or Akt downstream mTORC1 signaling inhibition did not fully block the IL-6 or LIF induction of protein synthesis. SOCS3 siRNA knockdown increased basal protein synthesis and extended the duration of the protein synthesis induction by IL-6 and LIF. These results demonstrate that either IL-6 or LIF can activate gp130-Akt signaling axis, which induces protein synthesis via mTORC1-independent mechanisms in cultured myotubes. However, IL-6- or LIF-induced SOCS3 negatively regulates the activation of myotube protein synthesis.
Collapse
Affiliation(s)
- Song Gao
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - J Larry Durstine
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Ho-Jin Koh
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Wayne E Carver
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina
| | - Norma Frizzell
- Department of Pharmacology, Physiology, and Neuronscience, School of Medicine, University of South Carolina, Columbia, South Carolina; and
| | - James A Carson
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina; .,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
44
|
Roselló-Díez A, Stephen D, Joyner AL. Altered paracrine signaling from the injured knee joint impairs postnatal long bone growth. eLife 2017; 6. [PMID: 28741471 PMCID: PMC5526667 DOI: 10.7554/elife.27210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/03/2017] [Indexed: 12/23/2022] Open
Abstract
Regulation of organ growth is a poorly understood process. In the long bones, the growth plates (GPs) drive elongation by generating a scaffold progressively replaced by bone. Although studies have focused on intrinsic GP regulation, classic and recent experiments suggest that local signals also modulate GP function. We devised a genetic mouse model to study extrinsic long bone growth modulation, in which injury is specifically induced in the left hindlimb, such that the right hindlimb serves as an internal control. Remarkably, when only mesenchyme cells surrounding postnatal GPs were killed, left bone growth was nevertheless reduced. GP signaling was impaired by altered paracrine signals from the knee joint, including activation of the injury response and, in neonates, dampened IGF1 production. Importantly, only the combined prevention of both responses rescued neonatal growth. Thus, we identified signals from the knee joint that modulate bone growth and could underlie establishment of body proportions. DOI:http://dx.doi.org/10.7554/eLife.27210.001 As bones grow, their size is carefully controlled and coordinated with the growth of the other organs in the body. The mechanisms that control organ size also help the body to recover from injury, and play a key role in controlling body size and proportions. Over the course of evolution, these mechanisms have likely changed to produce the distinct body sizes and proportions seen in humans and other animals. Despite their importance, it is not well understood how signals from both inside and outside an organ work together to regulate its size. In growth disorders this signaling goes wrong, which can lead to a person having unusual proportions such as a very short stature or having one leg shorter than the other. Currently, most growth disorders that affect leg proportions are treated with painful surgical procedures. Researchers would like to know how bone growth is affected by signals from the surrounding tissues because this could help them to develop new non-invasive treatments for these conditions. Long bones, for example those in the leg, grow from structures near their ends called growth plates. Roselló-Díez et al. have now engineered mice in which an injury shortly after birth caused cells in the knee in the rear left leg to die off. At the same time, the rear right leg of the mice developed as normal, allowing the growth of the two legs to be compared. Roselló-Díez et al. found that the left leg of these mice grew more slowly than the right leg, even though none of the cells in the growth plate of the left leg bone had been damaged. Further investigation revealed that this was because the injury caused an imbalance between the growth-promoting and growth-restricting signals that are produced by the fat pad and articular cartilage in the knee joint. Restoring the lost balance allowed the left leg bone to grow to a more normal length. In the future, boosting bone growth signals might provide a way to treat conditions like dwarfism or leg-length discrepancies. Understanding how different tissues influence body proportions could also help researchers to investigate how different animals evolved different body proportions. DOI:http://dx.doi.org/10.7554/eLife.27210.002
Collapse
Affiliation(s)
- Alberto Roselló-Díez
- Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Daniel Stephen
- Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Alexandra L Joyner
- Developmental Biology Program, Sloan Kettering Institute, New York, United States.,Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate Schoolof Medical Sciences, New York, United States
| |
Collapse
|
45
|
Kim M, Song K, Kim YS. Alantolactone Improves Prolonged Exposure of Interleukin-6-Induced Skeletal Muscle Inflammation Associated Glucose Intolerance and Insulin Resistance. Front Pharmacol 2017; 8:405. [PMID: 28706484 PMCID: PMC5489625 DOI: 10.3389/fphar.2017.00405] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 06/08/2017] [Indexed: 01/31/2023] Open
Abstract
The pro-inflammatory cytokine, Interleukin-6 (IL-6), has been proposed to be one of the mediators that link chronic inflammation to glucose intolerance and insulin resistance. Many studies have demonstrated the effects of IL-6 on insulin action in the skeletal muscle. However, few studies have investigated the effect of long-term treatment of IL-6, leading to glucose intolerance and insulin resistance. In the present study, we observed protective effects of alantolactone, a sesquiterpene lactone isolated from Inula helenium against glucose intolerance and insulin resistance induced by prolonged exposure of IL-6. Alantolactone has been reported to have anti-inflammatory and anti-cancer effects through IL-6-induced signal transducer and activator of transcription 3 (STAT3) signaling pathway. The relationship between IL-6 exposure and expression of toll-like receptor 4 (TLR4), involved in inflammation in the skeletal muscle, and the underlying mechanisms were investigated. We observed maximum dysregulation of glucose uptake after 40 ng/ml IL-6 induction for 24 h in L6 myotubes. Prolonged IL-6 exposure suppressed glucose uptake regulating alpha serine/threonine-protein kinase (AKT) phosphorylation; however, pretreatment with alantolactone activated AKT phosphorylation and improved glucose uptake. Alantolactone also attenuated IL-6-stimulated STAT3 phosphorylation, followed by an increase in expression of negative regulator suppressor of cytokine signaling 3 (SOCS3). Furthermore, IL-6-induced expression of pathogen recognition receptor, TLR4, was also suppressed by alantolactone pretreatment. Post-silencing of STAT3 using siRNA approach, IL-6-stimulated siRNA-STAT3 improved glucose uptake and suppressed TLR4 gene expression. Taken together, we propose that, as a STAT3 inhibitor, alantolactone, improves glucose regulation in the skeletal muscle by inhibiting IL-6-induced STAT3-SOCS3 signaling followed by inhibition of the TLR4 gene expression. Therefore, alantolactone can be a promising candidate for the treatment of inflammation-associated glucose intolerance and insulin resistance.
Collapse
Affiliation(s)
- Minjee Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National UniversitySeoul, South Korea
| | - Kwangho Song
- Natural Products Research Institute, College of Pharmacy, Seoul National UniversitySeoul, South Korea
| | - Yeong Shik Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National UniversitySeoul, South Korea
| |
Collapse
|
46
|
Zhang Q, Sun X, Xiao X, Zheng J, Li M, Yu M, Ping F, Wang Z, Qi C, Wang T, Wang X. Dietary Chromium Restriction of Pregnant Mice Changes the Methylation Status of Hepatic Genes Involved with Insulin Signaling in Adult Male Offspring. PLoS One 2017; 12:e0169889. [PMID: 28072825 PMCID: PMC5224989 DOI: 10.1371/journal.pone.0169889] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/22/2016] [Indexed: 12/14/2022] Open
Abstract
Maternal undernutrition is linked with an elevated risk of diabetes mellitus in offspring regardless of the postnatal dietary status. This is also found in maternal micro-nutrition deficiency, especial chromium which is a key glucose regulator. We investigated whether maternal chromium restriction contributes to the development of diabetes in offspring by affecting DNA methylation status in liver tissue. After being mated with control males, female weanling 8-week-old C57BL mice were fed a control diet (CON, 1.19 mg chromium/kg diet) or a low chromium diet (LC, 0.14 mg chromium/kg diet) during pregnancy and lactation. After weaning, some offspring were shifted to the other diet (CON-LC, or LC-CON), while others remained on the same diet (CON-CON, or LC-LC) for 29 weeks. Fasting blood glucose, serum insulin, and oral glucose tolerance test was performed to evaluate the glucose metabolism condition. Methylation differences in liver from the LC-CON group and CON-CON groups were studied by using a DNA methylation array. Bisulfite sequencing was carried out to validate the results of the methylation array. Maternal chromium limitation diet increased the body weight, blood glucose, and serum insulin levels. Even when switched to the control diet after weaning, the offspring also showed impaired glucose tolerance and insulin resistance. DNA methylation profiling of the offspring livers revealed 935 differentially methylated genes in livers of the maternal chromium restriction diet group. Pathway analysis identified the insulin signaling pathway was the main process affected by hypermethylated genes. Bisulfite sequencing confirmed that some genes in insulin signaling pathway were hypermethylated in livers of the LC-CON and LC-LC group. Accordingly, the expression of genes in insulin signaling pathway was downregulated. There findings suggest that maternal chromium restriction diet results in glucose intolerance in male offspring through alterations in DNA methylation which is associated with the insulin signaling pathway in the mice livers.
Collapse
Affiliation(s)
- Qian Zhang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaofang Sun
- Department of Endocrinology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- * E-mail:
| | - Jia Zheng
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming Li
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Miao Yu
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Fan Ping
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhixin Wang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Cuijuan Qi
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Tong Wang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaojing Wang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
47
|
Oh KJ, Lee DS, Kim WK, Han BS, Lee SC, Bae KH. Metabolic Adaptation in Obesity and Type II Diabetes: Myokines, Adipokines and Hepatokines. Int J Mol Sci 2016; 18:ijms18010008. [PMID: 28025491 PMCID: PMC5297643 DOI: 10.3390/ijms18010008] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/24/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
Obesity and type II diabetes are characterized by insulin resistance in peripheral tissues. A high caloric intake combined with a sedentary lifestyle is the leading cause of these conditions. Whole-body insulin resistance and its improvement are the result of the combined actions of each insulin-sensitive organ. Among the fundamental molecular mechanisms by which each organ is able to communicate and engage in cross-talk are cytokines or peptides which stem from secretory organs. Recently, it was reported that several cytokines or peptides are secreted from muscle (myokines), adipose tissue (adipokines) and liver (hepatokines) in response to certain nutrition and/or physical activity conditions. Cytokines exert autocrine, paracrine or endocrine effects for the maintenance of energy homeostasis. The present review is focused on the relationship and cross-talk amongst muscle, adipose tissue and the liver as secretory organs in metabolic diseases.
Collapse
Affiliation(s)
- Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Da Som Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Baek Soo Han
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| |
Collapse
|
48
|
Coope A, Torsoni AS, Velloso LA. MECHANISMS IN ENDOCRINOLOGY: Metabolic and inflammatory pathways on the pathogenesis of type 2 diabetes. Eur J Endocrinol 2016; 174:R175-87. [PMID: 26646937 DOI: 10.1530/eje-15-1065] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/07/2015] [Indexed: 12/17/2022]
Abstract
Obesity is the main risk factor for type 2 diabetes (T2D). Studies performed over the last 20 years have identified inflammation as the most important link between these two diseases. During the development of obesity, there is activation of subclinical inflammatory activity in tissues involved in metabolism and energy homeostasis. Intracellular serine/threonine kinases activated in response to inflammatory factors can catalyse the inhibitory phosphorylation of key proteins of the insulin-signalling pathway, leading to insulin resistance. Moreover, during the progression of obesity and insulin resistance, the pancreatic islets are also affected by inflammation, contributing to β-cell failure and leading to the onset of T2D. In this review, we will present the main mechanisms involved in the activation of obesity-associated metabolic inflammation and discuss potential therapeutic opportunities that can be developed to treat obesity-associated metabolic diseases.
Collapse
Affiliation(s)
- Andressa Coope
- Laboratory of Cell SignalingApplied Sciences FacultyUniversity of Campinas, 13084-970 Campinas, São Paulo, Brazil
| | - Adriana S Torsoni
- Laboratory of Cell SignalingApplied Sciences FacultyUniversity of Campinas, 13084-970 Campinas, São Paulo, Brazil
| | - Licio A Velloso
- Laboratory of Cell SignalingApplied Sciences FacultyUniversity of Campinas, 13084-970 Campinas, São Paulo, Brazil
| |
Collapse
|
49
|
Pinno J, Bongartz H, Klepsch O, Wundrack N, Poli V, Schaper F, Dittrich A. Interleukin-6 influences stress-signalling by reducing the expression of the mTOR-inhibitor REDD1 in a STAT3-dependent manner. Cell Signal 2016; 28:907-16. [PMID: 27094713 DOI: 10.1016/j.cellsig.2016.04.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 04/05/2016] [Accepted: 04/09/2016] [Indexed: 11/30/2022]
Abstract
Interleukin 6 (IL-6) is a pleiotropic cytokine and a strong activator of Mammalian Target of Rapamycin (mTOR). In contrast, mTOR activity is negatively regulated by Regulated in Development and DNA Damage Responses 1 (REDD1). Expression of REDD1 is induced by cellular stressors such as glucocorticoids and DNA damaging agents. We show that the expression of basal as well as stress-induced REDD1 is reduced by IL-6. The reduction of REDD1 expression by IL-6 is independent of proteasomal or caspase-mediated degradation of REDD1 protein. Instead, induction of REDD1 mRNA is reduced by IL-6. The regulation of REDD1 expression by IL-6 is independent of Phosphatidylinositide-3-Kinase (PI3K) and Mitogen-Activated Protein Kinase (MAPK) signalling but depends on the expression and activation of Signal Transducer and Activator of Transcription 3 (STAT3). Furthermore, the reduction of basal REDD1 expression by IL-6 correlates with IL-6-induced activation of mTOR signalling. Inhibition of STAT3 activation blocks IL-6-induced mTOR activation. In summary, we present a novel STAT3-dependent mechanism of both IL-6-induced activation of mTOR and IL-6-dependent reversion of stress-induced inhibition of mTOR activity.
Collapse
Affiliation(s)
- Jessica Pinno
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Hannes Bongartz
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Oliver Klepsch
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Nicole Wundrack
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Valeria Poli
- Molecular Biotechnology Center (MBC), Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy.
| | - Fred Schaper
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Anna Dittrich
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| |
Collapse
|
50
|
Huang J, Li L, Lian J, Schauer S, Vesely PW, Kratky D, Hoefler G, Lehner R. Tumor-Induced Hyperlipidemia Contributes to Tumor Growth. Cell Rep 2016; 15:336-48. [PMID: 27050512 PMCID: PMC4984953 DOI: 10.1016/j.celrep.2016.03.020] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 12/04/2015] [Accepted: 02/26/2016] [Indexed: 12/11/2022] Open
Abstract
The known link between obesity and cancer suggests an important interaction between the host lipid metabolism and tumorigenesis. Here, we used a syngeneic tumor graft model to demonstrate that tumor development influences the host lipid metabolism. BCR-Abl-transformed precursor B cell tumors induced hyperlipidemia by stimulating very low-density lipoprotein (VLDL) production and blunting VLDL and low-density lipoprotein (LDL) turnover. To assess whether tumor progression was dependent on tumor-induced hyperlipidemia, we utilized the VLDL production-deficient mouse model, carboxylesterase3/triacylglycerol hydrolase (Ces3/TGH) knockout mice. In Ces3/Tgh–/– tumor-bearing mice, plasma triglyceride and cholesterol levels were attenuated. Importantly tumor weight was reduced in Ces3/Tgh–/– mice. Mechanistically, reduced tumor growth in Ces3/Tgh–/– mice was attributed to reversal of tumor-induced PCSK9-mediated degradation of hepatic LDLR and decrease of LDL turnover. Our data demonstrate that tumor-induced hyperlipidemia encompasses a feed-forward loop that reprograms hepatic lipoprotein homeostasis in part by providing LDL cholesterol to support tumor growth.
Collapse
Affiliation(s)
- Jianfeng Huang
- Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Lena Li
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jihong Lian
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Silvia Schauer
- Institute of Pathology, Medical University of Graz, 8010 Graz, Austria
| | - Paul W Vesely
- Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; Institute of Molecular Biosciences, Karl Franzens University of Graz, 8010 Graz, Austria
| | - Dagmar Kratky
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Gerald Hoefler
- Institute of Pathology, Medical University of Graz, 8010 Graz, Austria.
| | - Richard Lehner
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|