1
|
Suleman S, Madsen AL, Ängquist LH, Schubert M, Linneberg A, Loos RJF, Hansen T, Grarup N. Genetic Underpinnings of Fasting and Oral Glucose-stimulated Based Insulin Sensitivity Indices. J Clin Endocrinol Metab 2024; 109:2754-2763. [PMID: 38635292 PMCID: PMC11479690 DOI: 10.1210/clinem/dgae275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/19/2024]
Abstract
CONTEXT Insulin sensitivity (IS) is an important factor in type 2 diabetes (T2D) and can be estimated by many different indices. OBJECTIVE We aimed to compare the genetic components underlying IS indices obtained from fasting and oral glucose-stimulated plasma glucose and serum insulin levels. METHODS We computed 21 IS indices, classified as fasting, OGTT0,120, and OGTT0,30,120 indices, using fasting and oral glucose tolerance test (OGTT) data in 2 cohorts. We used data from a family cohort (n = 313) to estimate the heritability and the genetic and phenotypic correlations of IS indices. The population cohort, Inter99 (n = 5343), was used to test for associations between IS indices and 426 genetic variants known to be associated with T2D. RESULTS Heritability estimates of IS indices ranged between 19% and 38%. Fasting and OGTT0,30,120 indices had high genetic (ρG) and phenotypic (ρP) pairwise correlations (ρG and ρP: 0.88 to 1) The OGTT0,120 indices displayed a wide range of pairwise correlations (ρG: 0.17-1.00 and ρP: 0.13-0.97). We identified statistically significant associations between IS indices and established T2D-associated variants. The PPARG rs11709077 variant was associated only with fasting indices and PIK3R rs4976033 only with OGTT0,30,120 indices. The variants in FAM63A/MINDY1, GCK, C2CD4A/B, and FTO loci were associated only with OGTT0,120 indices. CONCLUSION Even though the IS indices mostly share a common genetic background, notable differences emerged between OGTT0,120 indices. The fasting and OGTT-based indices have distinct associations with T2D risk variants. This work provides a basis for future large-scale genetic investigations into the differences between IS indices.
Collapse
Affiliation(s)
- Sufyan Suleman
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Anne L Madsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Lars H Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Mikkel Schubert
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen 2000, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Ruth J F Loos
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
2
|
Flores K, Almeida C, Arriaza K, Pena E, El Alam S. mTOR in the Development of Hypoxic Pulmonary Hypertension Associated with Cardiometabolic Risk Factors. Int J Mol Sci 2024; 25:11023. [PMID: 39456805 PMCID: PMC11508063 DOI: 10.3390/ijms252011023] [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: 09/12/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
The pathophysiology of pulmonary hypertension is complex and multifactorial. It is a disease characterized by increased pulmonary vascular resistance at the level due to sustained vasoconstriction and remodeling of the pulmonary arteries, which triggers an increase in the mean pulmonary artery pressure and subsequent right ventricular hypertrophy, which in some cases can cause right heart failure. Hypoxic pulmonary hypertension (HPH) is currently classified into Group 3 of the five different groups of pulmonary hypertensions, which are determined according to the cause of the disease. HPH mainly develops as a product of lung diseases, among the most prevalent causes of obstructive sleep apnea (OSA), chronic obstructive pulmonary disease (COPD), or hypobaric hypoxia due to exposure to high altitudes. Additionally, cardiometabolic risk factors converge on molecular mechanisms involving overactivation of the mammalian target of rapamycin (mTOR), which correspond to a central axis in the development of HPH. The aim of this review is to summarize the role of mTOR in the development of HPH associated with metabolic risk factors and its therapeutic alternatives, which will be discussed in this review.
Collapse
Affiliation(s)
| | | | - Karem Arriaza
- High Altitude Medicine Research Center (CEIMA), Arturo Prat University, Iquique 1110939, Chile; (K.F.); (C.A.); (E.P.); (S.E.A.)
| | | | | |
Collapse
|
3
|
Engin AB. Mechanism of Obesity-Related Lipotoxicity and Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:131-166. [PMID: 39287851 DOI: 10.1007/978-3-031-63657-8_5] [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 link between cellular exposure to fatty acid species and toxicity phenotypes remains poorly understood. However, structural characterization and functional profiling of human plasma free fatty acids (FFAs) analysis has revealed that FFAs are located either in the toxic cluster or in the cluster that is transcriptionally responsive to lipotoxic stress and creates genetic risk factors. Genome-wide short hairpin RNA screen has identified more than 350 genes modulating lipotoxicity. Hypertrophic adipocytes in obese adipose are both unable to expand further to store excess lipids in the diet and are resistant to the antilipolytic action of insulin. In addition to lipolysis, the inability of packaging the excess lipids into lipid droplets causes circulating fatty acids to reach toxic levels in non-adipose tissues. Deleterious effects of accumulated lipid in non-adipose tissues are known as lipotoxicity. Although triglycerides serve a storage function for long-chain non-esterified fatty acid and their products such as ceramide and diacylglycerols (DAGs), overloading of palmitic acid fraction of saturated fatty acids (SFAs) raises ceramide levels. The excess DAG and ceramide load create harmful effects on multiple organs and systems, inducing chronic inflammation in obesity. Thus, lipotoxic inflammation results in β cells death and pancreatic islets dysfunction. Endoplasmic reticulum stress stimuli induce lipolysis by activating cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) and extracellular signal-regulated kinase (Erk) 1/2 signaling in adipocytes. However, palmitic acid-induced endoplasmic reticulum stress-c-Jun N-terminal kinase (JNK)-autophagy axis in hypertrophic adipocytes is a pro-survival mechanism against endoplasmic reticulum stress and cell death induced by SFAs. Endoplasmic reticulum-localized acyl-coenzyme A (CoA): glycerol-3-phosphate acyltransferase (GPAT) enzymes are mediators of lipotoxicity, and inhibiting these enzymes has therapeutic potential for lipotoxicity. Lipotoxicity increases the number of autophagosomes, which engulf palmitic acid, and thus suppress the autophagic turnover. Fatty acid desaturation promotes palmitate detoxification and storages into triglycerides. As therapeutic targets of glucolipotoxicity, in addition to caloric restriction and exercise, there are four different pharmacological approaches, which consist of metformin, glucagon-like peptide 1 (GLP-1) receptor agonists, peroxisome proliferator-activated receptor-gamma (PPARγ) ligands thiazolidinediones, and chaperones are still used in clinical practice. Furthermore, induction of the brown fat-like phenotype with the mixture of eicosapentanoic acid and docosahexaenoic acid appears as a potential therapeutic application for treatment of lipotoxicity.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| |
Collapse
|
4
|
Domma AJ, Henderson LA, Goodrum FD, Moorman NJ, Kamil JP. Human cytomegalovirus attenuates AKT activity by destabilizing insulin receptor substrate proteins. J Virol 2023; 97:e0056323. [PMID: 37754763 PMCID: PMC10617551 DOI: 10.1128/jvi.00563-23] [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: 04/15/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023] Open
Abstract
IMPORTANCE Human cytomegalovirus (HCMV) requires inactivation of AKT to efficiently replicate, yet how AKT is shut off during HCMV infection has remained unclear. We show that UL38, an HCMV protein that activates mTORC1, is necessary and sufficient to destabilize insulin receptor substrate 1 (IRS1), a model insulin receptor substrate (IRS) protein. Degradation of IRS proteins in settings of excessive mTORC1 activity is an important mechanism for insulin resistance. When IRS proteins are destabilized, PI3K cannot be recruited to growth factor receptor complexes, and hence, AKT membrane recruitment, a rate limiting step in its activation, fails to occur. Despite its penchant for remodeling host cell signaling pathways, our results reveal that HCMV relies upon a cell-intrinsic negative regulatory feedback loop to inactivate AKT. Given that pharmacological inhibition of PI3K/AKT potently induces HCMV reactivation from latency, our findings also imply that the expression of UL38 activity must be tightly regulated within latently infected cells to avoid spontaneous reactivation.
Collapse
Affiliation(s)
- Anthony J. Domma
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
| | - Lauren A. Henderson
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
| | - Felicia D. Goodrum
- Department of Immunobiology, University of Arizona, Tucson, Arizona, USA
- Bio5 Institute, University of Arizona, Tucson, Arizona, USA
| | - Nathaniel J. Moorman
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jeremy P. Kamil
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
5
|
Okita K, Hikiji H, Koga A, Nagai-Yoshioka Y, Yamasaki R, Mitsugi S, Fujii W, Ariyoshi W. Ascorbic acid enhances chondrocyte differentiation of ATDC5 by accelerating insulin receptor signaling. Cell Biol Int 2023; 47:1737-1748. [PMID: 37381608 DOI: 10.1002/cbin.12067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 05/06/2023] [Accepted: 06/20/2023] [Indexed: 06/30/2023]
Abstract
Chondrogenesis is strictly regulated by several factors, including cytokines, hormones, and extracellular matrix proteins. Mouse teratocarcinoma-derived lineage cells, differentiate into chondrocytes in the presence of insulin. Although ascorbic acid promotes chondrogenic differentiation, the detailed regulative mechanisms underlying its role in chondrogenesis remain unclear. Therefore, in this study, we evaluated the effects of ascorbic acid on insulin-induced chondrogenic differentiation of ATDC5 cells and the underlying intracellular signaling. The results revealed that insulin-stimulated collagen deposition, matrix formation, calcification, and expression of chondrogenic differentiation marker genes in ATDC5 cells. This enhancement by insulin was amplified with the addition of ascorbic acid. Molecular analysis revealed that the activation of insulin-induced phosphoinositide 3-kinase (PI3K)/Akt signaling was enhanced in the presence of ascorbic acid. In contrast, Wnt/β-catenin signaling was suppressed during chondrocyte differentiation via upregulation of the Wnt agonist, secreted Frizzled-related protein 1 (sFRP-1) and 3 (sFRP-3). Notably, ascorbic acid upregulated the expression of insulin receptors and their substrates (IRS-1 and IRS-2). Furthermore, ascorbic acid reversed the suppression of IRS-1 and IRS-2 protein by insulin. These results indicate that ascorbic acid positively regulates the chondrogenic differentiation of ATDC5 cells via enhancement of insulin signaling. Our findings provide a substantial basis for further elucidation of the regulatory mechanisms of chondrocyte differentiation and the pathophysiology of OA, thus aiding in development of effective treatment strategies.
Collapse
Affiliation(s)
- Kaede Okita
- Department of Health Promotion, Division of Infections and Molecular Biology, Kyushu Dental University, Fukuoka, Japan
- School of Oral Health Sciences, Faculty of Dentistry, Kyushu Dental University, Fukuoka, Japan
| | - Hisako Hikiji
- School of Oral Health Sciences, Faculty of Dentistry, Kyushu Dental University, Fukuoka, Japan
| | - Ayaka Koga
- Department of Health Promotion, Division of Infections and Molecular Biology, Kyushu Dental University, Fukuoka, Japan
- School of Oral Health Sciences, Faculty of Dentistry, Kyushu Dental University, Fukuoka, Japan
| | - Yoshie Nagai-Yoshioka
- Department of Health Promotion, Division of Infections and Molecular Biology, Kyushu Dental University, Fukuoka, Japan
| | - Ryota Yamasaki
- Department of Health Promotion, Division of Infections and Molecular Biology, Kyushu Dental University, Fukuoka, Japan
| | - Sho Mitsugi
- Department of Science of Physical Functions, Division of Oral and Maxillofacial Surgery, Kyushu Dental University, Fukuoka, Japan
| | - Wataru Fujii
- School of Oral Health Sciences, Faculty of Dentistry, Kyushu Dental University, Fukuoka, Japan
| | - Wataru Ariyoshi
- Department of Health Promotion, Division of Infections and Molecular Biology, Kyushu Dental University, Fukuoka, Japan
| |
Collapse
|
6
|
Ignjatović Đ, Tovilović-Kovačević G, Mićić B, Tomić M, Djordjevic A, Macut D, Vojnović Milutinović D. Effects of early life overnutrition and hyperandrogenism on spatial learning and memory in a rat model of polycystic ovary syndrome. Horm Behav 2023; 153:105392. [PMID: 37295324 DOI: 10.1016/j.yhbeh.2023.105392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a complex disorder characterized by endocrine and metabolic abnormalities such as obesity and insulin resistance. PCOS is also associated with psychiatric disorders and cognitive impairment. The animal model of PCOS was induced by treating rats with 5α-dihydrotestosterone (5α-DHT) and additionally modified to induce adiposity by litter size reduction (LSR). Spatial learning and memory were assessed using the Barnes Maze test, and striatal markers of synaptic plasticity were analyzed. Striatal insulin signaling was estimated by the levels of insulin receptor substrate 1 (IRS1), its inhibitory phosphorylation at Ser307, and glycogen synthase kinase-3α/β (GSK3α/β) activity. Both LSR and DHT treatment significantly decreased striatal protein levels of IRS1, followed by increased GSK3α/β activity in small litters. Results of the behavioral study showed that LSR had a negative effect on learning rate and memory retention, whereas DHT treatment did not induce impairment in memory formation. While protein levels of synaptophysin, GAP43, and postsynaptic density protein 95 (PSD-95) were not altered by the treatments, DHT treatment induced an increase in phosphorylation of PSD-95 at Ser295 in both normal and small litters. This study revealed that LSR and DHT treatment suppressed insulin signaling by downregulating IRS1 in the striatum. However, DHT treatment did not have an adverse effect on learning and memory, probably due to compensatory elevation in pPSD-95-Ser295, which had a positive effect on synaptic strength. This implies that hyperandrogenemia in this setting does not represent a threat to spatial learning and memory, opposite to the effect of overnutrition-related adiposity.
Collapse
Affiliation(s)
- Đurđica Ignjatović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd., 11000 Belgrade, Serbia.
| | - Gordana Tovilović-Kovačević
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd., 11000 Belgrade, Serbia.
| | - Bojana Mićić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd., 11000 Belgrade, Serbia.
| | - Mirko Tomić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd., 11000 Belgrade, Serbia.
| | - Ana Djordjevic
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd., 11000 Belgrade, Serbia.
| | - Djuro Macut
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Dr Subotića 13, 11000 Belgrade, Serbia.
| | - Danijela Vojnović Milutinović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd., 11000 Belgrade, Serbia.
| |
Collapse
|
7
|
Zhang Y, Lan M, Liu C, Wang T, Liu C, Wu S, Meng Q. Islr regulates insulin sensitivity by interacting with Psma4 to control insulin receptor alpha levels in obese mice. Int J Biochem Cell Biol 2023; 159:106420. [PMID: 37116777 DOI: 10.1016/j.biocel.2023.106420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/30/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023]
Abstract
Insulin resistance is the leading cause of type 2 diabetes (T2D), and dysfunctional insulin receptor signaling is a major manifestation of this insulin resistance. In T2D, the corresponding insulin receptor levels are aberrantly down-regulated, which is one of the major factors underlying obesity-induced insulin resistance in adipose tissue. However, the precise mechanism of insulin receptor impairment in obese individuals remains unclear. In the current study, we established that immunoglobulin superfamily containing leucine-rich repeat (Islr) is highly expressed in adipocytes of mice fed a high-fat diet. We further demonstrated that Islr mediates the ubiquitin-independent proteasomal degradation of insulin receptor alpha (Insrα) by specifically interacting with proteasome subunit alpha type 4 (Psma4). Islr knockout increased the corresponding Insrα subunit levels and enhanced insulin sensitivity in adipocytes, ultimately improving systemic metabolism. Further, siRNA-mediated down-regulation of Islr expression in the white adipose tissue of obese mice increased insulin sensitivity. Overall, Islr regulates insulin sensitivity by interacting with Psma4 to control the ubiquitin-independent proteasomal degradation of Insrα in obese mice, indicating that Islr may be a potential therapeutic target for ameliorating insulin resistance.
Collapse
Affiliation(s)
- Yuying Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China; State Key Laboratories of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China
| | - Miaomiao Lan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China; State Key Laboratories of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China
| | - Chang Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China; State Key Laboratories of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China
| | - Tongtong Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China; State Key Laboratories of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China
| | - Chuncheng Liu
- State Key Laboratories of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China; The Institute of Bioengineering and Technology, Inner Mongolia University of Science and Technology, Baotou 014010, China
| | - Sen Wu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China; State Key Laboratories of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China
| | - Qingyong Meng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China; State Key Laboratories of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian, Beijing 100193, China.
| |
Collapse
|
8
|
Domma AJ, Goodrum FD, Moorman NJ, Kamil JP. Human cytomegalovirus attenuates AKT activity by destabilizing insulin receptor substrate proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537203. [PMID: 37131605 PMCID: PMC10153195 DOI: 10.1101/2023.04.17.537203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The phosphoinositide 3-kinase (PI3K)/AKT pathway plays crucial roles in cell viability and protein synthesis and is frequently co-opted by viruses to support their replication. Although many viruses maintain high levels of AKT activity during infection, other viruses, such as vesicular stomatitis virus and human cytomegalovirus (HCMV), cause AKT to accumulate in an inactive state. To efficiently replicate, HCMV requires FoxO transcription factors to localize to the infected cell nucleus (Zhang et. al. mBio 2022), a process directly antagonized by AKT. Therefore, we sought to investigate how HCMV inactivates AKT to achieve this. Subcellular fractionation and live cell imaging studies indicated that AKT failed to recruit to membranes upon serum-stimulation of infected cells. However, UV-inactivated virions were unable to render AKT non-responsive to serum, indicating a requirement for de novo viral gene expression. Interestingly, we were able to identify that UL38 (pUL38), a viral activator of mTORC1, is required to diminish AKT responsiveness to serum. mTORC1 contributes to insulin resistance by causing proteasomal degradation of insulin receptor substrate (IRS) proteins, such as IRS1, which are necessary for the recruitment of PI3K to growth factor receptors. In cells infected with a recombinant HCMV disrupted for UL38 , AKT responsiveness to serum is retained and IRS1 is not degraded. Furthermore, ectopic expression of UL38 in uninfected cells induces IRS1 degradation, inactivating AKT. These effects of UL38 were reversed by the mTORC1 inhibitor, rapamycin. Collectively, our results demonstrate that HCMV relies upon a cell-intrinsic negative feedback loop to render AKT inactive during productive infection.
Collapse
Affiliation(s)
- Anthony J. Domma
- Department of Microbiology and Immunology, LSU Health Sciences Center Shreveport, Shreveport Louisiana, USA
| | - Felicia D. Goodrum
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
- Bio5 Institute, University of Arizona, Tucson, AZ, USA
| | - Nathaniel J. Moorman
- Department of Microbiology and Immunology, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Jeremy P. Kamil
- Department of Microbiology and Immunology, LSU Health Sciences Center Shreveport, Shreveport Louisiana, USA
| |
Collapse
|
9
|
Akarsu E, Sayiner ZA, Balcı SO, Demirel C, Bozdag Z, Korkmaz M, Yılmaz I. Effects of antidiabetics and exercise therapy on suppressors of cytokine signaling-1, suppressors of cytokine signaling-3, and insulin receptor substrate-1 molecules in diabetes and obesity. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:112-118. [PMID: 36629649 PMCID: PMC9937604 DOI: 10.1590/1806-9282.20220856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 08/20/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Pathological destruction of insulin signaling molecules such as insulin receptor substrate, especially due to the increase in suppressors of cytokine signaling molecules, has been demonstrated in experimental diabetes. The contribution of suppressors of cytokine signaling proteins to the development of insulin resistance and the effects of antidiabetic drugs and exercise on suppressors of cytokine signaling proteins are not clearly known. METHODS A total of 48 Wistar albino adult male rats were divided into six groups: control group, obese group with diabetes, obese diabetic rats treated with metformin, obese diabetic rats treated with pioglitazone, obese diabetic rats treated with exenatide, and obese diabetic rats with applied exercise program. Immunohistochemical staining was performed in both the liver and adipose tissue. RESULTS There was a statistically significant decrease in suppressors of cytokine signaling-1, a decrease in suppressors of cytokine signaling-3, an increase in insulin receptor substrate-1, and a decrease in immunohistochemical staining in the obese group treated with metformin and exenatide compared to the obese group without treatment in the liver tissue (p<0.05). A statistically significant decrease in immunohistochemical staining of suppressors of cytokine signaling-1 and suppressors of cytokine signaling-3 was found in the obese group receiving exercise therapy compared to the obese group without treatment in visceral adipose tissue (p<0.05). Likewise, no significant immunohistochemistry staining was seen in diabetic obese groups. CONCLUSION Metformin or exenatide treatment could prevent the degradation of insulin receptor substrate-1 protein by reducing the effect of suppressors of cytokine signaling-1 and suppressors of cytokine signaling-3 proteins, especially in the liver tissue. In addition, exercise can play a role as a complementary therapy by reducing suppressors of cytokine signaling-1 and suppressors of cytokine signaling-3 proteins in visceral adipose tissue.
Collapse
Affiliation(s)
- Ersin Akarsu
- University of Gaziantep, Faculty of Medicine, Department of Endocrinology and Metabolism – Gaziantep, Turkey
| | - Zeynel Abidin Sayiner
- University of Gaziantep, Faculty of Medicine, Department of Endocrinology and Metabolism – Gaziantep, Turkey.,Corresponding author:
| | - Sibel Oğuzkan Balcı
- University of Gaziantep, Faculty of Medicine, Department of Medical Biology – Gaziantep, Turkey
| | - Can Demirel
- University of Gaziantep, Faculty of Medicine, Department of Biophysics – Gaziantep, Turkey
| | - Zehra Bozdag
- University of Gaziantep, Faculty of Medicine, Department of Pathology – Gaziantep, Turkey
| | - Murat Korkmaz
- University of Gaziantep, Faculty of Medicine, Department of Medical Biology – Gaziantep, Turkey
| | - Ibrahim Yılmaz
- University of Gaziantep, Faculty of Medicine, Department of Biophysics – Gaziantep, Turkey
| |
Collapse
|
10
|
Xu F, Zhang M, Wu H, Wang Y, Yang Y, Wang X. Study on the mechanism of lupenone for treating type 2 diabetes by integrating pharmacological evaluation and network pharmacology. PHARMACEUTICAL BIOLOGY 2022; 60:997-1010. [PMID: 35635284 PMCID: PMC9154797 DOI: 10.1080/13880209.2022.2067568] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
CONTEXT Lupenone (LUP) is the active ingredient of Musa basjoo Sieb. et Zucc. (Musaceae) with antidiabetes effects, but an unclear underlying mechanism of action. OBJECTIVE Animal experiments combined with network pharmacology were used to explore the mechanism of LUP for treating diabetes. MATERIALS AND METHODS Insulin resistance (IR) in male Sprague-Dawley rats with type 2 diabetic was induced using a high-fat diet and streptozotocin. The selected rats were divided into normal group, model group, positive group and LUP (2.0, 4.0 and 8.0 mg/kg) groups, and orally administrated twice daily with Tween 80, rosiglitazone or LUP. Fasting blood glucose (FBG), oxidative stress index, blood lipids and IR-related targets were detected. A network pharmacology analysis was performed. RESULTS Compared to the model group, LUP (8.0 mg/kg) significantly decreased the levels of FBG (22.3%), LEP (9.5%), HbA1c (14.9%) and MDA (12.3%), increased the ADPN (24.2%) levels and GSH-PX activity (12.4%) (p < 0.05), improved oxidative stress, lipid metabolism disorders and pancreas pathological changes, increased the mRNA and protein expression of InsR (3.7-fold and 1.3-fold), IRS-1 (3-fold and 2-fold), IRS-2 (2-fold and 1.6-fold), GLUT-4 (2-fold and 2.4-fold) in skeletal muscle and IRS-1 (6-fold and 1.6-fold), IRS-2 (5.8-fold and 1.5-fold), GLUT-4 (2.5-fold and 1.7-fold) and PPAR-γ (7-fold and 1.4-fold) in adipose tissue (p < 0.05). Network pharmacology analysis revealed that LUP improves IR by multiple targets and signal pathways. CONCLUSIONS The mechanism of LUP for treating diabetes is related to improving IR. LUP has the potential to be developed as a new drug for treating type 2 diabetes.
Collapse
Affiliation(s)
- Feng Xu
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, PR China
| | - Mei Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, PR China
| | - Hongmei Wu
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, PR China
| | - Yuanmin Wang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, PR China
| | - Ye Yang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, PR China
| | - Xiangpei Wang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, PR China
- College of Pharmacy, Guizhou Minzu University, Guiyang, PR China
- CONTACT Xiangpei Wang College of Pharmacy, Guizhou Minzu University, Huaxi District, Guizhou Province, Guiyang550025, PR China
| |
Collapse
|
11
|
Confettura AD, Cuboni E, Ammar MR, Jia S, Gomes GM, Yuanxiang P, Raman R, Li T, Grochowska KM, Ahrends R, Karpova A, Dityatev A, Kreutz MR. Neddylation-dependent protein degradation is a nexus between synaptic insulin resistance, neuroinflammation and Alzheimer's disease. Transl Neurodegener 2022; 11:2. [PMID: 34986876 PMCID: PMC8734066 DOI: 10.1186/s40035-021-00277-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The metabolic syndrome is a consequence of modern lifestyle that causes synaptic insulin resistance and cognitive deficits and that in interaction with a high amyloid load is an important risk factor for Alzheimer's disease. It has been proposed that neuroinflammation might be an intervening variable, but the underlying mechanisms are currently unknown. METHODS We utilized primary neurons to induce synaptic insulin resistance as well as a mouse model of high-risk aging that includes a high amyloid load, neuroinflammation, and diet-induced obesity to test hypotheses on underlying mechanisms. RESULTS We found that neddylation and subsequent activation of cullin-RING ligase complexes induced synaptic insulin resistance through ubiquitylation and degradation of the insulin-receptor substrate IRS1 that organizes synaptic insulin signaling. Accordingly, inhibition of neddylation preserved synaptic insulin signaling and rescued memory deficits in mice with a high amyloid load, which were fed with a 'western diet'. CONCLUSIONS Collectively, the data suggest that neddylation and degradation of the insulin-receptor substrate is a nodal point that links high amyloid load, neuroinflammation, and synaptic insulin resistance to cognitive decline and impaired synaptic plasticity in high-risk aging.
Collapse
Affiliation(s)
| | - Eleonora Cuboni
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Mohamed Rafeet Ammar
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Shaobo Jia
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany
| | - Guilherme M Gomes
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Otto Von Guericke University, 39120, Magdeburg, Germany
| | - PingAn Yuanxiang
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Rajeev Raman
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Tingting Li
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-e.V., 44227, Dortmund, Germany
| | - Katarzyna M Grochowska
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany.,Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Robert Ahrends
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-e.V., 44227, Dortmund, Germany.,Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090, Wien, Austria
| | - Anna Karpova
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Otto Von Guericke University, 39120, Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Otto Von Guericke University, 39120, Magdeburg, Germany.,Medical Faculty, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, 39118, Magdeburg, Germany. .,German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Otto Von Guericke University, 39120, Magdeburg, Germany. .,Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
12
|
Feng J, Zhou Y, Liao L, Yu L, Yuan P, Zhang J. Network Pharmacology and Transcriptomics Reveal the Mechanism of GuaLouQuMaiWan in Treatment of Type 2 Diabetes and Its Active Small Molecular Compound. J Diabetes Res 2022; 2022:2736504. [PMID: 36248223 PMCID: PMC9560855 DOI: 10.1155/2022/2736504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/18/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
The main pathophysiological abnormalities in type 2 diabetes (T2D) include pancreatic β-cell dysfunction and insulin resistance. Due to hyperglycemia, patients receive long-term treatment. However, side effects and drug tolerance usually lead to treatment failure. GuaLouQuMaiWan (GLQMW), a common traditional Chinese medicine (TCM) prescription, has positive effects on controlling blood sugar and improving quality of life, but the mechanism is still unclear. To decipher their molecular mechanisms, we used a novel computational systems pharmacology-based approach consisting of bioinformatics analysis, network pharmacology, and drug similarity comparison. We divided the participants into nondisease (ND), impaired glucose tolerance (IGT), and type 2 diabetes groups according to the WHO's recommendations for diabetes. By analyzing the gene expression profile of the ND-IGT-T2D (ND to IGT to T2D) process, we found that the function of downregulated genes in the whole process was mainly related to insulin secretion, while the upregulated genes were related to inflammation. Furthermore, other genes in the ND-IGT (ND to IGT) process are mainly related to inflammation and lipid metabolic disorders. We speculate that 17 genes with a consistent trend may play a key role in the process of ND-IGT-T2D. We further performed target prediction for 50 compounds in GLQMW that met the screening criteria and intersected the differentially expressed genes of the T2D process with the compounds of GLQMW; a total of 18 proteins proved potential targets for GLQMW. Among these, RBP4 is considerably related to insulin resistance. GO/KEGG enrichment analyses of the target genes of GLQMW showed enrichment in inflammation- and T2D therapy-related pathways. Based on the RDKit tool and the DrugBank database, we speculate that (-)-taxifolin, dialoside A_qt, spinasterol, isofucosterol, and 11,14-eicosadienoic acid can be used as potential drugs for T2D via molecular docking and drug similarity comparison.
Collapse
Affiliation(s)
- Jiahao Feng
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China 518000
| | - Yuheng Zhou
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China 510632
| | - Li Liao
- Chongqing Jiangjin District Hospital of Chinese Medicine, Chongqing, China 404100
| | - Liping Yu
- Shenzhen Hospital of Traditional Chinese Medicine, Shenzhen, China 518000
| | - Ping Yuan
- Tongren Hospital Shanghai Jiao Tong University, Shanghai, China 200000
| | - Jun Zhang
- School of Traditional Medicine, Jinan University, Guangzhou, China 510632
| |
Collapse
|
13
|
Gilbert M. Role of skeletal muscle lipids in the pathogenesis of insulin resistance of obesity and type 2 diabetes. J Diabetes Investig 2021; 12:1934-1941. [PMID: 34132491 PMCID: PMC8565406 DOI: 10.1111/jdi.13614] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity predisposes individuals to the development of insulin resistance, which is a risk factor for type 2 diabetes, and muscle plays a central role in this phenomenon. Insulin resistance is associated with: (i) a metabolic inflexibility characterized by a reduced impaired switching from free fatty acid (FA) to carbohydrate substrates; and (ii) an ectopic accumulation of triglyceride in skeletal muscle, generating a cellular "lipotoxicity", but triglyceride per se, does not contribute to insulin resistance ("athlete's paradox"). A large body of evidence supports the idea that a decreased mitochondrial capacity to oxidize FA leads to an accretion of intracellular triglyceride and an accumulation of acyl-CoAs, which are used to synthesize diacylglycerol and ceramide. These lipid derivatives activate serine kinases, leading to increase of insulin receptor substrate 1 serine phosphorylation, which impairs insulin signaling. A second model proposes that insulin resistance arises from an excessive mitochondrial FA oxidation. Studies have shown that the type of FA, unsaturated or saturated, is critical in the development of insulin resistance. It should be also stressed that FA oversupply activates inflammatory signals, induces endoplasmic reticulum stress, increases mitochondrial oxidative stress and influences the regulation of genes that contributes to impaired glucose metabolism. These cellular insults are thought to engage stress-sensitive serine kinases disrupting insulin signaling. In conclusion, reduced dietary lipid intake in association with physical exercise could be a therapeutic option to improve insulin sensitivity.
Collapse
Affiliation(s)
- Marc Gilbert
- CNRS UMR 8251 Bât. BuffonParis Diderot UniversityParisFrance
| |
Collapse
|
14
|
Jahantigh D, Mirani Sargazi F, Sargazi S, Saravani R, Ghazaey Zidanloo S, Heidari Nia M, Piri M. Relationship between Functional miR-143/145 Cluster Variants and Susceptibility to Type 2 Diabetes Mellitus: A Preliminary Case-Control Study and Bioinformatics Analyses. Endocr Res 2021; 46:129-139. [PMID: 33870836 DOI: 10.1080/07435800.2021.1914079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Purpose: To investigate the link between two variants (rs4705342 and rs4705343) in the promoter of the miR-143/145 cluster with Type 2 diabetes mellitus (T2DM) risk. Methods:A total of 1200 subjects were genotyped using the ARMS-PCR method. Results: The rs4705342 variant enhanced the risk of T2DM under codominant CC (OR = 3.24; 95% CI: 1.89-5.60), recessive TT+TC (OR = 3.02; 95% CI: 1.77-5.17), and dominant TC+CC (OR = 1.35; 95% CI: 1.08-1.71) genetic models. Individuals carrying the C allele of rs4705342 conferred a 1.43 fold increased risk of T2DM. As regards rs4705343, decreased risk of T2DM was observed under codominant TC (OR = 0.53; 95% CI: 0.42-0.67), over-dominant TT+CC (OR = 0.51; 95% CI: 0.40-0.64), and dominant TC+CC (OR = 0.59; 95% CI: 0.48-0.75) models. Haplotype analysis of the variants showed a 1.941-fold increased risk of T2DM regarding the C T combination. Significant associations were noticed between different haplotypes and lipid indices of T2DM patients. There were no notable changes in p-values after adjustment for BMI. Computational analysis revealed that miR143 and/or miR145 target important genes involved in glucose and lipid metabolism. Conclusions: Functional miR-143/145 variants might influence the risk of T2DM. Hence, clarifying the precise regulatory mechanisms of gene expression in the development of T2DM will significantly guide researchers to find a novel target for therapeutic intervention.
Collapse
Affiliation(s)
- Danial Jahantigh
- Department of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | - Fariba Mirani Sargazi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ramin Saravani
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | | | - Milad Heidari Nia
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Maryam Piri
- Diabetes Center, Ali Asghar Hospital, Zahedan University of Medical Sciences, Zahedan, IR Iran
| |
Collapse
|
15
|
Alipourfard I, Bakhtiyari S, Gheysarzadeh A, Di Renzo L, De Lorenzo A, Mikeladze D, Khamoushi A. The Key Role of Akt Protein Kinase in Metabolic-Inflammatory Pathways Cross-Talk: TNF-α Down-Regulation and Improving of Insulin Resistance in HepG2 Cell Line. Curr Mol Med 2021; 21:257-264. [PMID: 32338219 DOI: 10.2174/1566524020666200427102209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/02/2020] [Accepted: 04/12/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Elevation of plasma free fatty acids as a principal aspect of type 2 diabetes maintains etiologically insulin insensitivity in target cells. TNF-α inhibitory effects on key insulin signaling pathway elements remain to be verified in insulinresistant hepatic cells. Thus, TNF-α knockdown effects on the key elements of insulin signaling were investigated in the palmitate-induced insulin-resistant hepatocytes. The Akt serine kinase, a key protein of the insulin signaling pathway, phosphorylation was monitored to understand the TNF-α effect on probable enhancing of insulin resistance. METHODS Insulin-resistant HepG2 cells were produced using 0.5 mM palmitate treatment and shRNA-mediated TNF-α gene knockdown and its down-regulation confirmed using ELISA technique. Western blotting analysis was used to assess the Akt protein phosphorylation status. RESULTS Palmitate-induced insulin resistance caused TNF-α protein overexpression 1.2-, 2.78, and 2.25- fold as compared to the control cells at post-treatment times of 8 h, 16 h, and 24 h, respectively. In the presence of palmitate, TNF-α expression showed around 30% reduction in TNF-α knockdown cells as compared to normal cells. In the TNF-α down-regulated cell, Akt phosphorylation was approximately 62% more than control cells after treatment with 100 nM insulin in conjugation with 0.5 mM palmitate. CONCLUSIONS The obtained data demonstrated that TNF-α protein expression reduction improved insulin-stimulated Akt phosphorylation in the HepG2 cells and decreased lipidinduced insulin resistance of the diabetic hepatocytes.
Collapse
Affiliation(s)
- Iraj Alipourfard
- Institute of Chemical Biology, School of Natural Sciences and Engineering, Ilia State University, Tbilisi, Georgia
| | - Salar Bakhtiyari
- Department of Clinical Biochemistry, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Ali Gheysarzadeh
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Italy
| | - Antonio De Lorenzo
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Italy
| | - David Mikeladze
- Institute of Chemical Biology, School of Natural Sciences and Engineering, Ilia State University, Tbilisi, Georgia
| | | |
Collapse
|
16
|
Build-UPS and break-downs: metabolism impacts on proteostasis and aging. Cell Death Differ 2021; 28:505-521. [PMID: 33398091 PMCID: PMC7862225 DOI: 10.1038/s41418-020-00682-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/30/2022] Open
Abstract
Perturbation of metabolism elicits cellular stress which profoundly modulates the cellular proteome and thus protein homeostasis (proteostasis). Consequently, changes in the cellular proteome due to metabolic shift require adaptive mechanisms by molecular protein quality control. The mechanisms vitally controlling proteostasis embrace the entire life cycle of a protein involving translational control at the ribosome, chaperone-assisted native folding, and subcellular sorting as well as proteolysis by the proteasome or autophagy. While metabolic imbalance and proteostasis decline have been recognized as hallmarks of aging and age-associated diseases, both processes are largely considered independently. Here, we delineate how proteome stability is governed by insulin/IGF1 signaling (IIS), mechanistic target of Rapamycin (TOR), 5′ adenosine monophosphate-activated protein kinase (AMPK), and NAD-dependent deacetylases (Sir2-like proteins known as sirtuins). This comprehensive overview is emphasizing the regulatory interconnection between central metabolic pathways and proteostasis, indicating the relevance of shared signaling nodes as targets for future therapeutic interventions. ![]()
Collapse
|
17
|
Rocha M, Apostolova N, Diaz-Rua R, Muntane J, Victor VM. Mitochondria and T2D: Role of Autophagy, ER Stress, and Inflammasome. Trends Endocrinol Metab 2020; 31:725-741. [PMID: 32265079 DOI: 10.1016/j.tem.2020.03.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/08/2020] [Accepted: 03/05/2020] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes (T2D) is one of the main current threats to human health. Both T2D and its numerous clinical complications are related to mitochondrial dysfunction and oxidative stress. Over the past decade, great progress has been made in extending our knowledge about the signaling events regulated by mitochondria. However, the links among mitochondrial impairment, oxidative stress, autophagy, endoplasmic reticulum (ER) stress, and activation of the inflammasome still need to be clarified. In light of this deficit, we aim to provide a review of the existing literature concerning the complicated crosstalk between mitochondrial impairment, autophagy, ER stress, and the inflammasome in the molecular pathogenesis of T2D.
Collapse
Affiliation(s)
- Milagros Rocha
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain.
| | | | - Ruben Diaz-Rua
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Jordi Muntane
- Department of Pharmacology, University of Valencia, Valencia, Spain; Institute of Biomedicine of Seville (IBiS), University Hospital 'Virgen del Rocío'/CSIC/University of Seville, Seville, Spain; Department of General Surgery, University Hospital 'Virgen del Rocío'/CSIC/University of Seville/IBiS/CSIC/University of Seville, Spain
| | - Victor M Victor
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Department of Physiology, University of Valencia, Valencia, Spain.
| |
Collapse
|
18
|
Apostolova N, Iannantuoni F, Gruevska A, Muntane J, Rocha M, Victor VM. Mechanisms of action of metformin in type 2 diabetes: Effects on mitochondria and leukocyte-endothelium interactions. Redox Biol 2020; 34:101517. [PMID: 32535544 PMCID: PMC7296337 DOI: 10.1016/j.redox.2020.101517] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/13/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes (T2D) is a very prevalent, multisystemic, chronic metabolic disorder closely related to atherosclerosis and cardiovascular diseases. It is characterised by mitochondrial dysfunction and the presence of oxidative stress. Metformin is one of the safest and most effective anti-hyperglycaemic agents currently employed as first-line oral therapy for T2D. It has demonstrated additional beneficial effects, unrelated to its hypoglycaemic action, on weight loss and several diseases, such as cancer, cardiovascular disorders and metabolic diseases, including thyroid diseases. Despite the vast clinical experience gained over several decades of use, the mechanism of action of metformin is still not fully understood. This review provides an overview of the existing literature concerning the beneficial mitochondrial and vascular effects of metformin, which it exerts by diminishing oxidative stress and reducing leukocyte-endothelium interactions. Specifically, we describe the molecular mechanisms involved in metformin's effect on gluconeogenesis, its capacity to interfere with major metabolic pathways (AMPK and mTORC1), its action on mitochondria and its antioxidant effects. We also discuss potential targets for therapeutic intervention based on these molecular actions.
Collapse
Affiliation(s)
- Nadezda Apostolova
- Department of Pharmacology, University of Valencia - FISABIO (Foundation for the Promotion of Health and Biomedical Research in the Valencian Region), Valencia, Spain; CIBERehd (Biomedical Research Networking Centre on Hepatic and Digestive Diseases), Valencia, Spain.
| | - Francesca Iannantuoni
- Service of Endocrinology and Nutrition. University Hospital Doctor Peset, FISABIO, Valencia, Spain
| | - Aleksandra Gruevska
- Department of Pharmacology, University of Valencia - FISABIO (Foundation for the Promotion of Health and Biomedical Research in the Valencian Region), Valencia, Spain
| | - Jordi Muntane
- Institute of Biomedicine of Seville (IBiS), University Hospital "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Milagros Rocha
- CIBERehd (Biomedical Research Networking Centre on Hepatic and Digestive Diseases), Valencia, Spain; Service of Endocrinology and Nutrition. University Hospital Doctor Peset, FISABIO, Valencia, Spain
| | - Victor M Victor
- CIBERehd (Biomedical Research Networking Centre on Hepatic and Digestive Diseases), Valencia, Spain; Service of Endocrinology and Nutrition. University Hospital Doctor Peset, FISABIO, Valencia, Spain; Department of Physiology, University of Valencia, Valencia, Spain.
| |
Collapse
|
19
|
Chen Z, Tian R, She Z, Cai J, Li H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radic Biol Med 2020; 152:116-141. [PMID: 32156524 DOI: 10.1016/j.freeradbiomed.2020.02.025] [Citation(s) in RCA: 782] [Impact Index Per Article: 156.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide and is strongly associated with the presence of oxidative stress. Disturbances in lipid metabolism lead to hepatic lipid accumulation, which affects different reactive oxygen species (ROS) generators, including mitochondria, endoplasmic reticulum, and NADPH oxidase. Mitochondrial function adapts to NAFLD mainly through the downregulation of the electron transport chain (ETC) and the preserved or enhanced capacity of mitochondrial fatty acid oxidation, which stimulates ROS overproduction within different ETC components upstream of cytochrome c oxidase. However, non-ETC sources of ROS, in particular, fatty acid β-oxidation, appear to produce more ROS in hepatic metabolic diseases. Endoplasmic reticulum stress and NADPH oxidase alterations are also associated with NAFLD, but the degree of their contribution to oxidative stress in NAFLD remains unclear. Increased ROS generation induces changes in insulin sensitivity and in the expression and activity of key enzymes involved in lipid metabolism. Moreover, the interaction between redox signaling and innate immune signaling forms a complex network that regulates inflammatory responses. Based on the mechanistic view described above, this review summarizes the mechanisms that may account for the excessive production of ROS, the potential mechanistic roles of ROS that drive NAFLD progression, and therapeutic interventions that are related to oxidative stress.
Collapse
Affiliation(s)
- Ze Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Ruifeng Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Zhigang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China; Basic Medical School, Wuhan University, Wuhan, 430071, PR China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, PR China
| | - Jingjing Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China; Basic Medical School, Wuhan University, Wuhan, 430071, PR China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, PR China.
| |
Collapse
|
20
|
Siddiqui JA, Pothuraju R, Jain M, Batra SK, Nasser MW. Advances in cancer cachexia: Intersection between affected organs, mediators, and pharmacological interventions. Biochim Biophys Acta Rev Cancer 2020; 1873:188359. [PMID: 32222610 DOI: 10.1016/j.bbcan.2020.188359] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/10/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023]
Abstract
Advanced cancer patients exhibit cachexia, a condition characterized by a significant reduction in the body weight predominantly from loss of skeletal muscle and adipose tissue. Cachexia is one of the major causes of morbidity and mortality in cancer patients. Decreased food intake and multi-organ energy imbalance in cancer patients worsen the cachexia syndrome. Cachectic cancer patients have a low tolerance for chemo- and radiation therapies and also have a reduced quality of life. The presence of tumors and the current treatment options for cancer further exacerbate the cachexia condition, which remains an unmet medical need. The onset of cachexia involves crosstalk between different organs leading to muscle wasting. Recent advancements in understanding the molecular mechanisms of skeletal muscle atrophy/hypertrophy and adipose tissue wasting/browning provide a platform for the development of new targeted therapies. Therefore, a better understanding of this multifactorial disorder will help to improve the quality of life of cachectic patients. In this review, we summarize the metabolic mediators of cachexia, their molecular functions, affected organs especially with respect to muscle atrophy and adipose browning and then discuss advanced therapeutic approaches to cancer cachexia.
Collapse
Affiliation(s)
- Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ramesh Pothuraju
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Mohd W Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
21
|
Hentschel A, Ahrends R. Developing a Robust Assay to Monitor and Quantify Key Players of Metabolic Pathways during Adipogenesis by Targeted Proteomics. Proteomics 2020; 20:e1900141. [PMID: 32196961 DOI: 10.1002/pmic.201900141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 03/09/2020] [Indexed: 12/22/2022]
Abstract
Targeted data acquisition using nano liquid chromatrography (nano-LC) coupled mass spectrometry is an emerging approach when there is a need to quantify proteins with high accuracy, sensitivity, and reproducibility. Nevertheless, creating assays meeting all those criteria still remains a laborious task, especially when investigating low abundant proteins and small concentration changes. In this work a targeted data acquisition workflow is developed reducing time and effort to target and investigate key players of metabolic pathways during the process of adipocyte differentiation. This leads to accurate and sensitive quantification of proteins involved in the synthesis of fatty acids, glycerolipids, glycerophospholipids, sphingolipids, the production of energy and reduction equivalents. Additionally low abundant signaling molecules part of the peroxisome proliferator-activated receptor gamma (PPARγ) and insulin signaling pathway with ≈400 for the insulin receptor substrate and 1100 copies per cell for PPARγ are determined.
Collapse
Affiliation(s)
- Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bioanalytics, Standartisation, Otto-Hahn-Straße 6b, Dortmund, D-44227, Germany
| | - Robert Ahrends
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bioanalytics, Standartisation, Otto-Hahn-Straße 6b, Dortmund, D-44227, Germany.,Institute of Analytical Chemistry, University of Vienna, Währinger str. 38, Vienna, A-1090, Austria
| |
Collapse
|
22
|
Pan ZQ. Cullin-RING E3 Ubiquitin Ligase 7 in Growth Control and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1217:285-296. [PMID: 31898234 PMCID: PMC8343956 DOI: 10.1007/978-981-15-1025-0_17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
CRL7Fbxw8 is an E3 ubiquitin ligase complex, containing cullin7 (CUL7) as a scaffold, the F-box protein Fbxw8 as a substrate receptor, the Skp1 adaptor, and the ROC1/Rbx1 RING finger protein for working with E2 enzyme to facilitate ubiquitin transfer. This chapter provides an update on studies linking CRL7Fbxw8 to hereditary human growth retardation disease, as at least 64 cul7 germ line mutations were found in patients with autosomal recessive 3-M syndrome. CRL7Fbxw8 interacts with two additional 3-M associated proteins OBSL1 and CCDC8, leading to subcellular localization of the E3 complex to regions including plasma membrane, centrosome, and Golgi. At least ten mammalian cellular proteins were identified or implicated as CRL7Fbxw8 substrates. Discussion focuses on the possible impact of CRL7Fbxw8-mediated proteolytic or non-proteolytic pathways in growth control and cancer.
Collapse
Affiliation(s)
- Zhen-Qiang Pan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
23
|
Tian S, Jia W, Lu M, Zhao J, Sun X. Dual-specificity tyrosine phosphorylation-regulated kinase 1A ameliorates insulin resistance in neurons by up-regulating IRS-1 expression. J Biol Chem 2019; 294:20164-20176. [PMID: 31723029 PMCID: PMC6937568 DOI: 10.1074/jbc.ra119.010809] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/10/2019] [Indexed: 11/06/2022] Open
Abstract
Insulin resistance in the brain is a pathological mechanism that is shared between Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM). Although aberrant expression and phosphorylation of insulin receptor substrate 1 (IRS-1) contribute to insulin resistance, the underlying mechanism remains elusive. In this study, we used several approaches, including adeno-associated virus-based protein overexpression, immunoblotting, immunoprecipitation, immunohistochemistry, and in situ proximal ligation assays, to investigate the function of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) in IRS-1 regulation and the downstream insulin signaling in neurons. We found that DYRK1A overexpression up-regulated IRS-1 expression by slowing turnover of the IRS-1 protein. We further observed that DYRK1A directly interacted with IRS-1 and phosphorylated IRS-1's multiple serine residues. Of note, DYRK1A and IRS-1 were coordinately up-regulated in the prefrontal cortex of db/db mice brain. Furthermore, DYRK1A overexpression ameliorated chronic high insulin-induced insulin resistance in SH-SY5Y cells as well as in primary rat neurons. These findings suggest that DYRK1A protects against insulin resistance in the brain by elevating IRS-1 expression.
Collapse
Affiliation(s)
- Shijiao Tian
- Department of Neurology, Qilu Hospital of Shandong University, No. 107 Wenhuaxi Rd., 250012 Jinan, China
- Brain Research Institute, Qilu Hospital of Shandong University, No. 107 Wenhuaxi Rd., 250012 Jinan, China
| | - Wenming Jia
- NHC Key Laboratory of Otorhinolaryngology, Chinese Ministry of Health, Qilu Hospital of Shandong University, No. 44 Wenhuaxi Rd., 250012 Jinan, China
| | - Mei Lu
- Department of Geriatrics, Qilu Hospital of Shandong University, No. 107 Wenhuaxi Rd., 250012 Jinan, China
| | - Juan Zhao
- NHC Key Laboratory of Otorhinolaryngology, Chinese Ministry of Health, Qilu Hospital of Shandong University, No. 44 Wenhuaxi Rd., 250012 Jinan, China
| | - Xiulian Sun
- Brain Research Institute, Qilu Hospital of Shandong University, No. 107 Wenhuaxi Rd., 250012 Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission, Qilu Hospital of Shandong University, No. 107 West Wenhua Rd., Jinan, 250012 Shandong Province, China
| |
Collapse
|
24
|
Astaxanthin: A Potential Mitochondrial-Targeted Antioxidant Treatment in Diseases and with Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3849692. [PMID: 31814873 PMCID: PMC6878783 DOI: 10.1155/2019/3849692] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 08/30/2019] [Indexed: 12/23/2022]
Abstract
Oxidative stress is characterized by an imbalance between prooxidant and antioxidant species, leading to macromolecular damage and disruption of redox signaling and cellular control. It is a hallmark of various diseases including metabolic syndrome, chronic fatigue syndrome, neurodegenerative, cardiovascular, inflammatory, and age-related diseases. Several mitochondrial defects have been considered to contribute to the development of oxidative stress and known as the major mediators of the aging process and subsequent age-associated diseases. Thus, mitochondrial-targeted antioxidants should prevent or slow down these processes and prolong longevity. This is the reason why antioxidant treatments are extensively studied and newer and newer compounds with such an effect appear. Astaxanthin, a xanthophyll carotenoid, is the most abundant carotenoid in marine organisms and is one of the most powerful natural compounds with remarkable antioxidant activity. Here, we summarize its antioxidant targets, effects, and benefits in diseases and with aging.
Collapse
|
25
|
Frendo-Cumbo S, Jaldin-Fincati JR, Coyaud E, Laurent EMN, Townsend LK, Tan JMJ, Xavier RJ, Pillon NJ, Raught B, Wright DC, Brumell JH, Klip A. Deficiency of the autophagy gene ATG16L1 induces insulin resistance through KLHL9/KLHL13/CUL3-mediated IRS1 degradation. J Biol Chem 2019; 294:16172-16185. [PMID: 31515271 DOI: 10.1074/jbc.ra119.009110] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/29/2019] [Indexed: 12/23/2022] Open
Abstract
Connections between deficient autophagy and insulin resistance have emerged, however, the mechanism through which reduced autophagy impairs insulin-signaling remains unknown. We examined mouse embryonic fibroblasts lacking Atg16l1 (ATG16L1 KO mouse embryonic fibroblasts (MEFs)), an essential autophagy gene, and observed deficient insulin and insulin-like growth factor-1 signaling. ATG16L1 KO MEFs displayed reduced protein content of insulin receptor substrate-1 (IRS1), pivotal to insulin signaling, whereas IRS1myc overexpression recovered downstream insulin signaling. Endogenous IRS1 protein content and insulin signaling were restored in ATG16L1 KO mouse embryonic fibroblasts (MEF) upon proteasome inhibition. Through proximity-dependent biotin identification (BioID) and co-immunoprecipitation, we found that Kelch-like proteins KLHL9 and KLHL13, which together form an E3 ubiquitin (Ub) ligase complex with cullin 3 (CUL3), are novel IRS1 interactors. Expression of Klhl9 and Klhl13 was elevated in ATG16L1 KO MEFs and siRNA-mediated knockdown of Klhl9, Klhl13, or Cul3 recovered IRS1 expression. Moreover, Klhl13 and Cul3 knockdown increased insulin signaling. Notably, adipose tissue of high-fat fed mice displayed lower Atg16l1 mRNA expression and IRS1 protein content, and adipose tissue KLHL13 and CUL3 expression positively correlated to body mass index in humans. We propose that ATG16L1 deficiency evokes insulin resistance through induction of Klhl9 and Klhl13, which, in complex with Cul3, promote proteasomal IRS1 degradation.
Collapse
Affiliation(s)
- Scott Frendo-Cumbo
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | - Etienne Coyaud
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2C1, Canada
| | - Estelle M N Laurent
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2C1, Canada
| | - Logan K Townsend
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Joel M J Tan
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Ramnik J Xavier
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| | - Nicolas J Pillon
- Departments of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden 171 77
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2C1, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - David C Wright
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - John Hunter Brumell
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada .,Department of Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,SickKids IBD Centre, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Amira Klip
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada .,Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
26
|
Gorgisen G, Hapil FZ, Yilmaz O, Cetin Z, Pehlivanoglu S, Ozbudak IH, Erdogan A, Ozes ON. Identification of novel mutations of Insulin Receptor Substrate 1 (IRS1) in tumor samples of non-small cell lung cancer (NSCLC): Implications for aberrant insulin signaling in development of cancer. Genet Mol Biol 2019; 42:15-25. [PMID: 30807634 PMCID: PMC6428125 DOI: 10.1590/1678-4685-gmb-2017-0307] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 06/13/2018] [Indexed: 02/08/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related death, and NSCLC constitutes nearly 85%-90% of all cases. The IRS proteins function as adaptors and transmit signals from multiple receptors. Upon binding of insulin to the insulin receptor (IR), IRS1 is phosphorylated at several YXXM motifs creating docking sites for the binding of PI3Kp85, which activates AKT kinase. Therefore, we thought that gain of function mutantions of IRS1 could be related to development of lung cancer. In line with this, we wanted determine whether the IRS1 gene was mutated in the coding regions surrounding YXXM motifs. We sequenced the coding regions surrounding YXXM motifs of IRS1 using tumor samples of 42 NSCLC patients and 40 matching controls and found heterozygote p.S668T mutation in nine of 42 samples and four of nine also had the p.D674H mutation. We generated IRS1 expression vectors harboring p.S668T, p.D674H and double mutants. Expression of the mutants differentially affected insulin-induced phosphorylation of IRS1, AKT, ERK, and STAT3. Also, our mutants induced proliferation, glucose uptake, inhibited the migration of 293T cells and affected the responsiveness of the cells to cisplatin and radiation. Our results suggest that these novel mutations play a role in the phenotype of lung cancer.
Collapse
Affiliation(s)
| | | | - Ozlem Yilmaz
- Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Zafer Cetin
- Faculty of Medicine, Sanko University, Gaziantep, Turkey
| | | | | | | | | |
Collapse
|
27
|
Park S, Kim YS, Kim DY, So I, Jeon JH. PI3K pathway in prostate cancer: All resistant roads lead to PI3K. Biochim Biophys Acta Rev Cancer 2018; 1870:198-206. [PMID: 30300679 DOI: 10.1016/j.bbcan.2018.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/13/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022]
|
28
|
Rovira-Llopis S, Apostolova N, Bañuls C, Muntané J, Rocha M, Victor VM. Mitochondria, the NLRP3 Inflammasome, and Sirtuins in Type 2 Diabetes: New Therapeutic Targets. Antioxid Redox Signal 2018; 29:749-791. [PMID: 29256638 DOI: 10.1089/ars.2017.7313] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Type 2 diabetes mellitus and hyperglycemia can lead to the development of comorbidities such as atherosclerosis and microvascular/macrovascular complications. Both type 2 diabetes and its complications are related to mitochondrial dysfunction and oxidative stress. Type 2 diabetes is also a chronic inflammatory condition that leads to inflammasome activation and the release of proinflammatory mediators, including interleukins (ILs) IL-1β and IL-18. Moreover, sirtuins are energetic sensors that respond to metabolic load, which highlights their relevance in metabolic diseases, such as type 2 diabetes. Recent Advances: Over the past decade, great progress has been made in clarifying the signaling events regulated by mitochondria, inflammasomes, and sirtuins. Nod-like receptor family pyrin domain containing 3 (NLRP3) is the best characterized inflammasome, and the generation of oxidant species seems to be critical for its activation. NLRP3 inflammasome activation and altered sirtuin levels have been observed in type 2 diabetes. Critical Issue: Despite increasing evidence of the relationship between the NLRP3 inflammasome, mitochondrial dysfunction, and oxidative stress and of their participation in type 2 diabetes physiopathology, therapeutic strategies to combat type 2 diabetes that target NLRP3 inflammasome and sirtuins are yet to be consolidated. FUTURE DIRECTIONS In this review article, we attempt to provide an overview of the existing literature concerning the crosstalk between mitochondrial impairment and the inflammasome, with particular attention to cellular and mitochondrial redox metabolism and the potential role of the NLRP3 inflammasome and sirtuins in the pathogenesis of type 2 diabetes. In addition, we discuss potential targets for therapeutic intervention based on these molecular interactions. Antioxid. Redox Signal. 29, 749-791.
Collapse
Affiliation(s)
- Susana Rovira-Llopis
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Nadezda Apostolova
- 2 Department of Pharmacology, University of Valencia , Valencia, Spain
- 4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
| | - Celia Bañuls
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Jordi Muntané
- 3 Department of General Surgery, Hospital University "Virgen del Rocío"/IBiS/CSIC/University of Seville , Seville, Spain
- 4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
| | - Milagros Rocha
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
- 4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
| | - Victor M Victor
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
- 4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
- 5 Department of Physiology, University of Valencia , Valencia, Spain
| |
Collapse
|
29
|
Rabiee A, Krüger M, Ardenkjær-Larsen J, Kahn CR, Emanuelli B. Distinct signalling properties of insulin receptor substrate (IRS)-1 and IRS-2 in mediating insulin/IGF-1 action. Cell Signal 2018; 47:1-15. [PMID: 29550500 DOI: 10.1016/j.cellsig.2018.03.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 12/21/2022]
Abstract
Insulin/IGF-1 action is driven by a complex and highly integrated signalling network. Loss-of-function studies indicate that the major insulin/IGF-1 receptor substrate (IRS) proteins, IRS-1 and IRS-2, mediate different biological functions in vitro and in vivo, suggesting specific signalling properties despite their high degree of homology. To identify mechanisms contributing to the differential signalling properties of IRS-1 and IRS-2 in the mediation of insulin/IGF-1 action, we performed comprehensive mass spectrometry (MS)-based phosphoproteomic profiling of brown preadipocytes from wild type, IRS-1-/- and IRS-2-/- mice in the basal and IGF-1-stimulated states. We applied stable isotope labeling by amino acids in cell culture (SILAC) for the accurate quantitation of changes in protein phosphorylation. We found ~10% of the 6262 unique phosphorylation sites detected to be regulated by IGF-1. These regulated sites included previously reported substrates of the insulin/IGF-1 signalling pathway, as well as novel substrates including Nuclear Factor I X and Semaphorin-4B. In silico prediction suggests the protein kinase B (PKB), protein kinase C (PKC), and cyclin-dependent kinase (CDK) as the main mediators of these phosphorylation events. Importantly, we found preferential phosphorylation patterns depending on the presence of either IRS-1 or IRS-2, which was associated with specific sets of kinases involved in signal transduction downstream of these substrates such as PDHK1, MAPK3, and PKD1 for IRS-1, and PIN1 and PKC beta for IRS-2. Overall, by generating a comprehensive phosphoproteomic profile from brown preadipocyte cells in response to IGF-1 stimulation, we reveal both common and distinct insulin/IGF-1 signalling events mediated by specific IRS proteins.
Collapse
Affiliation(s)
- Atefeh Rabiee
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Marcus Krüger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jacob Ardenkjær-Larsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - C Ronald Kahn
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Brice Emanuelli
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark.
| |
Collapse
|
30
|
Wardhana DA, Ikeda K, Barinda AJ, Nugroho DB, Qurania KR, Yagi K, Miyata K, Oike Y, Hirata KI, Emoto N. Family with sequence similarity 13, member A modulates adipocyte insulin signaling and preserves systemic metabolic homeostasis. Proc Natl Acad Sci U S A 2018; 115:1529-1534. [PMID: 29386390 PMCID: PMC5816206 DOI: 10.1073/pnas.1720475115] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Adipose tissue dysfunction is causally implicated in the impaired metabolic homeostasis associated with obesity; however, detailed mechanisms underlying dysregulated adipocyte functions in obesity remain to be elucidated. Here we searched for genes that provide a previously unknown mechanism in adipocyte metabolic functions and identified family with sequence similarity 13, member A (Fam13a) as a factor that modifies insulin signal cascade in adipocytes. Fam13a was highly expressed in adipose tissue, predominantly in mature adipocytes, and its expression was substantially reduced in adipose tissues of obese compared with lean mice. We revealed that Fam13a accentuated insulin signaling by recruiting protein phosphatase 2A with insulin receptor substrate 1 (IRS1), leading to protection of IRS1 from proteasomal degradation. We further demonstrated that genetic loss of Fam13a exacerbated obesity-related metabolic disorders, while targeted activation of Fam13a in adipocytes ameliorated it in association with altered adipose tissue insulin sensitivity in mice. Our data unveiled a previously unknown mechanism in the regulation of adipocyte insulin signaling by Fam13a and identified its significant role in systemic metabolic homeostasis, shedding light on Fam13a as a pharmacotherapeutic target to treat obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Donytra Arby Wardhana
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Higashinada, 658-8558 Kobe, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo, 6500017 Kobe, Japan
| | - Koji Ikeda
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Higashinada, 658-8558 Kobe, Japan;
| | - Agian Jeffilano Barinda
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Higashinada, 658-8558 Kobe, Japan
| | - Dhite Bayu Nugroho
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Higashinada, 658-8558 Kobe, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo, 6500017 Kobe, Japan
| | - Kikid Rucira Qurania
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Higashinada, 658-8558 Kobe, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo, 6500017 Kobe, Japan
| | - Keiko Yagi
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Higashinada, 658-8558 Kobe, Japan
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, Chuo-ku, 860-8556 Kumamoto, Japan
- Department of Immunology, Allergy and Vascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Chuo-ku, 860-8556 Kumamoto, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, Chuo-ku, 860-8556 Kumamoto, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo, 6500017 Kobe, Japan
| | - Noriaki Emoto
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Higashinada, 658-8558 Kobe, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo, 6500017 Kobe, Japan
| |
Collapse
|
31
|
Exercise training reverses the negative effects of chronic L-arginine supplementation on insulin sensitivity. Life Sci 2017; 191:17-23. [DOI: 10.1016/j.lfs.2017.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/24/2017] [Accepted: 10/01/2017] [Indexed: 12/14/2022]
|
32
|
Abstract
Enlarged fat cells in obese adipose tissue diminish capacity to store fat and are resistant to the anti-lipolytic effect of insulin. Insulin resistance (IR)-associated S-nitrosylation of insulin-signaling proteins increases in obesity. In accordance with the inhibition of insulin-mediated anti-lipolytic action, plasma free fatty acid (FFA) levels increase. Additionally, endoplasmic reticulum stress stimuli induce lipolysis by activating cyclic adenosine monophosphate/Protein kinase A (cAMP/PKA) and extracellular signal-regulated kinase ½ (ERK1/2) signaling in adipocytes. Failure of packaging of excess lipid into lipid droplets causes chronic elevation of circulating fatty acids, which can reach to toxic levels within non-adipose tissues. Deleterious effects of lipid accumulation in non-adipose tissues are known as lipotoxicity. In fact, triglycerides may also serve a storage function for long-chain non-esterified fatty acids and their products such as ceramides and diacylglycerols (DAGs). Thus, excess DAG, ceramide and saturated fatty acids in obesity can induce chronic inflammation and have harmful effect on multiple organs and systems. In this context, chronic adipose tissue inflammation, mitochondrial dysfunction and IR have been discussed within the scope of lipotoxicity.
Collapse
|
33
|
Di Meo S, Iossa S, Venditti P. Skeletal muscle insulin resistance: role of mitochondria and other ROS sources. J Endocrinol 2017; 233:R15-R42. [PMID: 28232636 DOI: 10.1530/joe-16-0598] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
Abstract
At present, obesity is one of the most important public health problems in the world because it causes several diseases and reduces life expectancy. Although it is well known that insulin resistance plays a pivotal role in the development of type 2 diabetes mellitus (the more frequent disease in obese people) the link between obesity and insulin resistance is yet a matter of debate. One of the most deleterious effects of obesity is the deposition of lipids in non-adipose tissues when the capacity of adipose tissue is overwhelmed. During the last decade, reduced mitochondrial function has been considered as an important contributor to 'toxic' lipid metabolite accumulation and consequent insulin resistance. More recent reports suggest that mitochondrial dysfunction is not an early event in the development of insulin resistance, but rather a complication of the hyperlipidemia-induced reactive oxygen species (ROS) production in skeletal muscle, which might promote mitochondrial alterations, lipid accumulation and inhibition of insulin action. Here, we review the literature dealing with the mitochondria-centered mechanisms proposed to explain the onset of obesity-linked IR in skeletal muscle. We conclude that the different pathways leading to insulin resistance may act synergistically because ROS production by mitochondria and other sources can result in mitochondrial dysfunction, which in turn can further increase ROS production leading to the establishment of a harmful positive feedback loop.
Collapse
Affiliation(s)
- Sergio Di Meo
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Susanna Iossa
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Paola Venditti
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| |
Collapse
|
34
|
mTORC1 Plays an Important Role in Skeletal Development by Controlling Preosteoblast Differentiation. Mol Cell Biol 2017; 37:MCB.00668-16. [PMID: 28069737 DOI: 10.1128/mcb.00668-16] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 01/03/2017] [Indexed: 01/15/2023] Open
Abstract
The mammalian target of rapamycin complex 1 (mTORC1) is activated by extracellular factors that control bone accrual. However, the direct role of this complex in osteoblast biology remains to be determined. To investigate this question, we disrupted mTORC1 function in preosteoblasts by targeted deletion of Raptor (Rptor) in Osterix-expressing cells. Deletion of Rptor resulted in reduced limb length that was associated with smaller epiphyseal growth plates in the postnatal skeleton. Rptor deletion caused a marked reduction in pre- and postnatal bone accrual, which was evident in skeletal elements derived from both intramembranous and endochondrial ossification. The decrease in bone accrual, as well as the associated increase in skeletal fragility, was due to a reduction in osteoblast function. In vitro, osteoblasts derived from knockout mice display a reduced osteogenic potential, and an assessment of bone-developmental markers in Rptor knockout osteoblasts revealed a transcriptional profile consistent with an immature osteoblast phenotype suggesting that osteoblast differentiation was stalled early in osteogenesis. Metabolic labeling and an assessment of cell size of Rptor knockout osteoblasts revealed a significant decrease in protein synthesis, a major driver of cell growth. These findings demonstrate that mTORC1 plays an important role in skeletal development by regulating mRNA translation during preosteoblast differentiation.
Collapse
|
35
|
Disruption of the Phosphate Transporter Pit1 in Hepatocytes Improves Glucose Metabolism and Insulin Signaling by Modulating the USP7/IRS1 Interaction. Cell Rep 2016; 16:2736-2748. [PMID: 27568561 DOI: 10.1016/j.celrep.2016.08.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 06/02/2016] [Accepted: 08/03/2016] [Indexed: 01/07/2023] Open
Abstract
The liver plays a central role in whole-body lipid and glucose homeostasis. Increasing dietary fat intake results in increased hepatic fat deposition, which is associated with a risk for development of insulin resistance and type 2 diabetes. In this study, we demonstrate a role for the phosphate inorganic transporter 1 (PiT1/SLC20A1) in regulating metabolism. Specific knockout of Pit1 in hepatocytes significantly improved glucose tolerance and insulin sensitivity, enhanced insulin signaling, and decreased hepatic lipogenesis. We identified USP7 as a PiT1 binding partner and demonstrated that Pit1 deletion inhibited USP7/IRS1 dissociation upon insulin stimulation. This prevented IRS1 ubiquitination and its subsequent proteasomal degradation. As a consequence, delayed insulin negative feedback loop and sustained insulin signaling were observed. Moreover, PiT1-deficient mice were protected against high-fat-diet-induced obesity and diabetes. Our findings indicate that PiT1 has potential as a therapeutic target in the context of metabolic syndrome, obesity, and diabetes.
Collapse
|
36
|
Buta C, Benabou E, Lequoy M, Régnault H, Wendum D, Meratbene F, Chettouh H, Aoudjehane L, Conti F, Chrétien Y, Scatton O, Rosmorduc O, Praz F, Fartoux L, Desbois-Mouthon C. Heregulin-1ß and HER3 in hepatocellular carcinoma: status and regulation by insulin. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:126. [PMID: 27514687 PMCID: PMC4982118 DOI: 10.1186/s13046-016-0402-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 07/28/2016] [Indexed: 02/06/2023]
Abstract
Background The heregulin-1ß/HER3-driven pathway is implicated in several epithelial malignancies and its blockade is currently undergoing clinical investigation. Paradoxically, the status and the regulation of this pathway is poorly known in hepatocellular carcinoma (HCC). Methods Using 85 HCC obtained after tumour resection, heregulin-1ß and HER3 expression was evaluated by real-time RT-PCR, ELISA and/or immunohistochemistry. Statistics were performed to analyze associations between gene expression and clinicopathological parameters. The effects of insulin on the heregulin-1ß/HER3 pathway was investigated in four HCC cell lines. Results HER3 mRNA was upregulated in 52 % of tumours, while heregulin-1ß mRNA was downregulated in 82 %. Hepatitis B and C viral infections were respectively associated with high and low HER3 mRNA expression. No association was seen between neither HER3 or heregulin-1ß mRNA and prognostic factors, survival or recurrence. Immunohistochemistry showed predominant cytoplasmic staining of HER3 in tumours but the staining was nonreproducible. HER3 mRNA and protein levels were not correlated in liver tissues. In HCC cells, insulin promoted HER3 proteasomal degradation and inhibited heregulin-1ß stimulation of cell migration. HER3 and insulin receptor co-immunoprecipitated in these cells. The loss of insulin receptor expression by RNA interference sensitized cells to heregulin-1ß-induced AKT phosphorylation. Conclusions Autocrine heregulin-1ß loop is uncommon in HCC and HER3 mRNA expression is differentially influenced by hepatitis viruses. Insulin is a negative regulator of HER3 protein expression and function in HCC cells. Altogether these data may explain why HER3 and heregulin-1ß expression have no prognostic value and suggest that HCC patients are unlikely to derive benefit from HER3-targeted monotherapies. Electronic supplementary material The online version of this article (doi:10.1186/s13046-016-0402-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Corina Buta
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France
| | - Eva Benabou
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France
| | - Marie Lequoy
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France.,Department of Hepatology, AP-HP, Saint-Antoine Hospital, F-75012, Paris, France
| | - Hélène Régnault
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France.,Department of Hepatology, AP-HP, Pitié-Salpétrière Hospital, F-75013, Paris, France
| | - Dominique Wendum
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France.,Department of Pathology, AP-HP, Saint-Antoine Hospital, F-75012, Paris, France
| | - Fatiha Meratbene
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France.,Histomorphology Platform, UMS 30 Lumic, F-75012, Paris, France
| | - Hamza Chettouh
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France
| | - Lynda Aoudjehane
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France.,Human HepCell, Saint-Antoine Hospital, F-75012, Paris, France
| | - Filomena Conti
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France.,Human HepCell, Saint-Antoine Hospital, F-75012, Paris, France.,Department of Hepatobiliary Surgery and Liver Transplantation, AP-HP, Pitié-Salpétrière Hospital, F-75013, Paris, France
| | - Yves Chrétien
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France
| | - Olivier Scatton
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France.,Department of Hepatobiliary Surgery and Liver Transplantation, AP-HP, Pitié-Salpétrière Hospital, F-75013, Paris, France
| | - Olivier Rosmorduc
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France.,Department of Hepatology, AP-HP, Pitié-Salpétrière Hospital, F-75013, Paris, France
| | - Françoise Praz
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France
| | - Laetitia Fartoux
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France.,Department of Hepatology, AP-HP, Pitié-Salpétrière Hospital, F-75013, Paris, France
| | - Christèle Desbois-Mouthon
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Saint-Antoine Research Center, 34 rue Crozatier, F-75012, Paris, France.
| |
Collapse
|
37
|
Jean-Charles PY, Freedman NJ, Shenoy SK. Chapter Nine - Cellular Roles of Beta-Arrestins as Substrates and Adaptors of Ubiquitination and Deubiquitination. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:339-69. [PMID: 27378762 DOI: 10.1016/bs.pmbts.2016.04.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
β-Arrestin1 and β-arrestin2 are homologous adaptor proteins that are ubiquitously expressed in mammalian cells. They belong to a four-member family of arrestins that regulate the vast family of seven-transmembrane receptors that couple to heterotrimeric G proteins (7TMRs or GPCRs), and that modulate 7TMR signal transduction. β-Arrestins were originally identified in the context of signal inhibition via the 7TMRs because they competed with and thereby blocked G protein coupling to 7TMRs. Currently, in addition to their role as desensitizers of signaling, β-arrestins are appreciated as multifunctional adaptors that mediate trafficking and signal transduction of not only 7TMRs, but a growing list of additional receptors, ion channels, and nonreceptor proteins. β-Arrestins' interactions with their multifarious partners are based on their dynamic conformational states rather than particular domain-domain interactions. β-Arrestins adopt activated conformations upon 7TMR association. In addition, β-arrestins undergo various posttranslational modifications that are choreographed by activated 7TMRs, including phosphorylation, ubiquitination, acetylation, nitrosylation, and SUMOylation. Ubiquitination of β-arrestins is critical for their high-affinity interaction with 7TMRs as well as with endocytic adaptor proteins and signaling kinases. β-Arrestins also function as critical adaptors for ubiquitination and deubiquitination of various cellular proteins, and thereby affect the longevity of signal transducers and the intensity of signal transmission.
Collapse
Affiliation(s)
- P-Y Jean-Charles
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, North Carolina, United States
| | - N J Freedman
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, North Carolina, United States; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States
| | - S K Shenoy
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, North Carolina, United States; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States.
| |
Collapse
|
38
|
Copps KD, Hançer NJ, Qiu W, White MF. Serine 302 Phosphorylation of Mouse Insulin Receptor Substrate 1 (IRS1) Is Dispensable for Normal Insulin Signaling and Feedback Regulation by Hepatic S6 Kinase. J Biol Chem 2016; 291:8602-17. [PMID: 26846849 DOI: 10.1074/jbc.m116.714915] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Indexed: 12/30/2022] Open
Abstract
Constitutive activation of the mammalian target of rapamycin complex 1 and S6 kinase (mTORC1→ S6K) attenuates insulin-stimulated Akt activity in certain tumors in part through "feedback" phosphorylation of the upstream insulin receptor substrate 1 (IRS1). However, the significance of this mechanism for regulating insulin sensitivity in normal tissue remains unclear. We investigated the function of Ser-302 in mouse IRS1, the major site of its phosphorylation by S6K in vitro, through genetic knock-in of a serine-to-alanine mutation (A302). Although insulin rapidly stimulated feedback phosphorylation of Ser-302 in mouse liver and muscle, homozygous A302 mice (A/A) and their knock-in controls (S/S) exhibited similar glucose homeostasis and muscle insulin signaling. Furthermore, both A302 and control primary hepatocytes from which Irs2 was deleted showed marked inhibition of insulin-stimulated IRS1 tyrosine phosphorylation and PI3K binding after emetine treatment to raise intracellular amino acids and activate mTORC1 → S6K signaling. To specifically activate mTORC1 in mouse tissue, we deleted hepatic Tsc1 using Cre adenovirus. Although it moderately decreased IRS1/PI3K association and Akt phosphorylation in liver, Tsc1 deletion failed to cause glucose intolerance or promote hyperinsulinemia in mixed background A/A or S/S mice. Moreover, Tsc1 deletion failed to stimulate phospho-Ser-302 or other putative S6K sites within IRS1, whereas ribosomal S6 protein was constitutively phosphorylated. Following acute Tsc1 deletion from hepatocytes, Akt phosphorylation, but not IRS1/PI3K association, was rapidly restored by treatment with the mTORC1 inhibitor rapamycin. Thus, within the hepatic compartment, mTORC1 → S6K signaling regulates Akt largely through IRS-independent means with little effect upon physiologic insulin sensitivity.
Collapse
Affiliation(s)
- Kyle D Copps
- From the Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Nancy J Hançer
- From the Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Wei Qiu
- From the Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Morris F White
- From the Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
39
|
Zhao H, Huang X, Jiao J, Zhang H, Liu J, Qin W, Meng X, Shen T, Lin Y, Chu J, Li J. Protein phosphatase 4 (PP4) functions as a critical regulator in tumor necrosis factor (TNF)-α-induced hepatic insulin resistance. Sci Rep 2015; 5:18093. [PMID: 26666849 PMCID: PMC4678300 DOI: 10.1038/srep18093] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/11/2015] [Indexed: 12/27/2022] Open
Abstract
Protein phosphatase 4 (PP4) was shown to participate in multiple cellular processes, including DNA damage response, cell cycle and embryo development. Recent studies demonstrated a looming role of PP4 in glucose metabolism. However, whether PP4 is involved in hepatic insulin resistance remains poorly understood. The objective of this study was to estimate the role of PP4 in tumor necrosis factor (TNF)-α-induced hepatic insulin resistance. db/db mice and TNF-α-treated C57BL/6J mice were used as hepatic insulin resistance animal models. In vitro models were established in both HepG2 cells and primary hepatocytes by TNF-α treatment. We found that increased expression and activity of PP4 occurred in the livers of db/db mice and TNF-α-induced hepatic insulin resistance both in vitro and in vivo. Actually, PP4 silencing and suppression of PP4 activity ameliorated TNF-α-induced hepatic insulin resistance, whereas over-expression of PP4 caused insulin resistance. We then further investigated the prodiabetic mechanism of PP4 in TNF-α-induced insulin resistance. We found that PP4 formed a complex with IRS-1 to promote phosphorylation of IRS-1 on serine 307 via JNK activation and reduce the expression of IRS-1. Thus, PP4 is an important regulator in inflammatory related insulin resistance.
Collapse
Affiliation(s)
- Hongye Zhao
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.,The Key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| | - Xiuqing Huang
- The Key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| | - Juan Jiao
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.,The Key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| | - Hangxiang Zhang
- The Key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| | - Jin Liu
- College of Life Sciences, Beijing Normal University, 100875, P.R. China
| | - Weiwei Qin
- The Key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| | - Xiangyu Meng
- The Key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| | - Tao Shen
- The Key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| | - Yajun Lin
- The Key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| | - Jiaojiao Chu
- The Key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| | - Jian Li
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.,The Key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| |
Collapse
|
40
|
Kaempferol alleviates insulin resistance via hepatic IKK/NF-κB signal in type 2 diabetic rats. Int Immunopharmacol 2015; 28:744-50. [PMID: 26263168 DOI: 10.1016/j.intimp.2015.07.018] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 06/10/2015] [Accepted: 07/14/2015] [Indexed: 02/06/2023]
|
41
|
Kim KM, Lee KS, Lee GY, Jin H, Durrance ES, Park HS, Choi SH, Park KS, Kim YB, Jang HC, Lim S. Anti-diabetic efficacy of KICG1338, a novel glycogen synthase kinase-3β inhibitor, and its molecular characterization in animal models of type 2 diabetes and insulin resistance. Mol Cell Endocrinol 2015; 409:1-10. [PMID: 25802191 DOI: 10.1016/j.mce.2015.03.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/10/2015] [Accepted: 03/17/2015] [Indexed: 02/06/2023]
Abstract
Selective inhibition of glycogen synthase kinase-3 (GSK3) has been targeted as a novel therapeutic strategy for diabetes mellitus. We investigated the anti-diabetic efficacy and molecular mechanisms of KICG1338 (2-(4-fluoro-phenyl)-3H-imidazo[4,5-b]pyridine-7-carboxylic acid(4-methyl-pyridin-3-yl)-amide), a GSK3β inhibitor, in three animal models: Otsuka Long-Evans Tokushima Fatty (OLETF) rats, leptin receptors-deficient db/db mice, and diet-induced obese (DIO) mice. Biochemical parameters including glucose tolerance tests and gene expressions associated with glucose metabolism were investigated. Glucose excursion decreased significantly by KICG1338-treated OLETF rats, accompanied by increase in insulin receptor substrate-1 and glucose transporter (GLUT)-4 expressions in muscle and decreased GLUT-2 expression in liver. Glucose-lowering effects were similarly observed in KICG1338-treated db/db and DIO mice. KICG1338 treatment increased adiponectin levels and decreased TNF-α levels. KICG1338 therapy also led to greater β-cell preservation and less hepatic fat infiltration with decreased expressions of genes involved in inflammation and endoplasmic reticulum stress. These data demonstrate anti-diabetic efficacy of KICG1338, a novel GSK3β inhibitor.
Collapse
Affiliation(s)
- Kyoung Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Kuy-Sook Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Gha Young Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hyunjin Jin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Eunice Sung Durrance
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ho Seon Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung Hee Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine Seoul National University, Seoul, Republic of Korea
| | - Young-Bum Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine Seoul National University, Seoul, Republic of Korea; Division of Endocrinology, Metabolism and Diabetes, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Hak Chul Jang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
| |
Collapse
|
42
|
Yuan Y, Wang X, Lu X, Marunaka Y, Wang X. Effect of Coptidis Rhizoma extracts in a water-based solution on insulin resistance in 3T3-L1 adipocytes. Biomed Res 2015; 35:321-7. [PMID: 25355439 DOI: 10.2220/biomedres.35.321] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The present study was designed to investigate effects and molecular mechanisms of Coptidis Rhizoma extracts (CRE) on the improvement of insulin resistance induced by tumor necrosis factor-α (TNF-α) in adipocytes. We examined whether CRE administration could directly influence the insulin resistance in 3T3-L1 adipocytes. Potential roles of CRE in glucose consumption, mRNA expression of peroxisome proliferators activated receptor (PPAR-γ), expression of insulin receptor substrate-1 (IRS-1) protein, and phosphorylation of IRS-1 Ser307 were also investigated in the present study. Our data demonstrated that TNF-α significantly reduced levels of glucose consumption and IRS-1 protein expression, while TNF-α increased the phosphorylation of IRS-1 Ser307 in adipocytes 24 h after the challenge, suggesting that TNF-α induced a condition with the occurrence of insulin resistance. Those alterations induced by TNF-α were prevented and the mRNA expression of PPAR-γ was up-regulated by the administration of CRE. Thus, our results indicate that CRE can be used to prevent from the TNF-α-induced insulin resistance through PPAR-γ pathways.
Collapse
Affiliation(s)
- Yi Yuan
- Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine
| | | | | | | | | |
Collapse
|
43
|
Kim MJ, Rangasamy S, Shim Y, Song JM. Cell lysis-free quantum dot multicolor cellular imaging-based mechanism study for TNF-α-induced insulin resistance. J Nanobiotechnology 2015; 13:4. [PMID: 25623542 PMCID: PMC4310030 DOI: 10.1186/s12951-015-0064-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/07/2015] [Indexed: 12/25/2022] Open
Abstract
Background TNF-α is an inflammatory cytokine that plays an important role in insulin resistance observed in obesity and chronic inflammation. Many cellular components involved in insulin signaling cascade are known to be inhibited by TNF-α. Insulin receptor substrate (IRS)-1 is one of the major targets in TNF-α-induced insulin resistance. The serine phosphorylation of IRS-1 enables the inhibition of insulin signaling. Until now, many studies have been conducted to investigate the mechanism of TNF-α-induced insulin resistance based on Western blot. Intracellular protein kinase crosstalk is commonly encountered in inflammation-associated insulin resistance. The crosstalk among the signaling molecules obscures the precise role of kinases in insulin resistance. We have developed a cell lysis-free quantum dots (QDots) multicolor cellular imaging to identify the biochemical role of multiple kinases (p38, JNK, IKKβ, IRS1ser, IRS1tyr, GSK3β, and FOXO1) in inflammation-associated insulin resistance pathway with a single assay in one run. QDot-antibody conjugates were used as nanoprobes to simultaneously monitor the activation/deactivation of the above seven intracellular kinases in HepG2 cells. The effect of the test compounds on the suppression of TNF-α-induced insulin resistance was validated through kinase monitoring. Aspirin, indomethacin, cinnamic acid, and amygdalin were tested. Results Through the measurement of the glycogen level in HepG2 cell treated with TNF-α, it was found that aspirin and indomethacin increased glycogen levels by almost two-fold compared to amygdalin and cinnamic acid. The glucose production assay proved that cinnamic acid was much more efficient in suppressing glucose production, compared with MAP kinase inhibitors and non-steroidal anti-inflammatory drugs. QDot multicolor cellular imaging demonstrated that amygdalin and cinnamic acid selectively acted via the JNK1-dependent pathway to suppress the inflammation-induced insulin resistance and improve insulin sensitivity. Conclusion The regulatory function of multiple kinases could be monitored concurrently at the cellular level. The developed cellular imaging assay provides a unique platform for the understanding of inflammation and insulin resistance signaling pathways in type II diabetes mellitus and how they regulate each other. The results showed that amygdalin and cinnamic acid inhibit serine phosphorylation of IRS-1 through targeting JNK serine kinase and enhance insulin sensitivity.
Collapse
Affiliation(s)
| | | | | | - Joon Myong Song
- College of Pharmacy, Seoul National University, Seoul 151-742, South Korea.
| |
Collapse
|
44
|
Sasaki T, Kuroko M, Sekine S, Matsui S, Kikuchi O, Susanti VY, Kobayashi M, Tanaka Y, Yuasa T, Kitamura T. Overexpression of insulin receptor partially improves obese and diabetic phenotypes in db/db mice. Endocr J 2015; 62:787-96. [PMID: 26096452 DOI: 10.1507/endocrj.ej15-0255] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the major health concern among the world. Several treatment options for T2DM are in clinical use, including injecting insulin, promoting insulin secretion by insulin secretagogues, and improving insulin sensitivity by insulin sensitizers. However, increasing the amount of insulin receptor in insulin-target tissues has not been explored. In order to test the efficacy of insulin receptor overexpression for improving glucose control, we established a transgenic mouse line expressing human insulin receptor (INSR). We analyzed, growth, energy balance, and glucose control of INSR-overexpressing db/db mice (INSR; db/db), which we produced by mating INSR transgenic mice with db/db mice, a genetic model of obesity due to insufficient leptin signaling. Compared to db/db mice, INSR; db/db mice were rescued from hyperphagia and obesity, leading to improved blood glucose levels. Unexpectedly, however, INSR; db/db mice presented with stunted growth, accompanied by decreased plasma levels of free IGF1 and IGFBP-3, indicating the down-regulation of GH/IGF1 axis. These phenotypes were observed in INSR; db/db mice but not in INSR littermates. Meanwhile, bone defects observed in db/db male mice were not rescued. Moreover, improved blood glucose was not accompanied by improved insulin sensitivity. Therefore, overexpression of insulin receptor improves obese and diabetic phenotypes in db/db mice, with consequences on growth.
Collapse
Affiliation(s)
- Tsutomu Sasaki
- Laboratory of Metabolic Signal, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ishii M, Maeda A, Tani S, Akagawa M. Palmitate induces insulin resistance in human HepG2 hepatocytes by enhancing ubiquitination and proteasomal degradation of key insulin signaling molecules. Arch Biochem Biophys 2015; 566:26-35. [DOI: 10.1016/j.abb.2014.12.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/20/2014] [Accepted: 12/07/2014] [Indexed: 01/05/2023]
|
46
|
Hakuno F, Fukushima T, Yoneyama Y, Kamei H, Ozoe A, Yoshihara H, Yamanaka D, Shibano T, Sone-Yonezawa M, Yu BC, Chida K, Takahashi SI. The Novel Functions of High-Molecular-Mass Complexes Containing Insulin Receptor Substrates in Mediation and Modulation of Insulin-Like Activities: Emerging Concept of Diverse Functions by IRS-Associated Proteins. Front Endocrinol (Lausanne) 2015; 6:73. [PMID: 26074875 PMCID: PMC4443775 DOI: 10.3389/fendo.2015.00073] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/25/2015] [Indexed: 12/25/2022] Open
Abstract
Insulin-like peptides, such as insulin-like growth factors (IGFs) and insulin, induce a variety of bioactivities, such as growth, differentiation, survival, increased anabolism, and decreased catabolism in many cell types and in vivo. In general, IGFs or insulin bind to IGF-I receptor (IGF-IR) or insulin receptor (IR), activating the receptor tyrosine kinase. Insulin receptor substrates (IRSs) are known to be major substrates of receptor kinases, mediating IGF/insulin signals to direct bioactivities. Recently, we discovered that IRSs form high-molecular-mass complexes (referred to here as IRSomes) even without IGF/insulin stimulation. These complexes contain proteins (referred to here as IRSAPs; IRS-associated proteins), which modulate tyrosine phosphorylation of IRSs by receptor kinases, control IRS stability, and determine intracellular localization of IRSs. In addition, in these complexes, we found not only proteins that are involved in RNA metabolism but also RNAs themselves. Thus, IRSAPs possibly contribute to modulation of IGF/insulin bioactivities. Since it is established that disorder of modulation of insulin-like activities causes various age-related diseases including cancer, we could propose that the IRSome is an important target for treatment of these diseases.
Collapse
Affiliation(s)
- Fumihiko Hakuno
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshiaki Fukushima
- Laboratory of Biomedical Chemistry, Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Biological Sciences, Faculty of Bioscience and Biotechnology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Yosuke Yoneyama
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyasu Kamei
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Atsufumi Ozoe
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidehito Yoshihara
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Daisuke Yamanaka
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takashi Shibano
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Meri Sone-Yonezawa
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Bu-Chin Yu
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuhiro Chida
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichiro Takahashi
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- *Correspondence: Shin-Ichiro Takahashi, Laboratory of Cell Regulation, Departments of Animal Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan,
| |
Collapse
|
47
|
Yonamine CY, Teixeira SS, Campello RS, Gerlinger-Romero F, Rodrigues CF, Guimarães-Ferreira L, Machado UF, Nunes MT. Beta hydroxy beta methylbutyrate supplementation impairs peripheral insulin sensitivity in healthy sedentary Wistar rats. Acta Physiol (Oxf) 2014; 212:62-74. [PMID: 24962220 DOI: 10.1111/apha.12336] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/28/2014] [Accepted: 06/19/2014] [Indexed: 12/13/2022]
Abstract
AIM Investigate, in healthy sedentary rats, the potential mechanisms involved on the effects of beta hydroxy beta methylbutyrate (HMB) supplementation upon the glycaemic homeostasis, by evaluating the insulin sensitivity in liver, skeletal muscle, and white adipose tissue. METHODS Rats were supplemented with either beta hydroxy beta methylbutyrate (320 mg kg(-1) BW) or saline by gavage for 4 weeks. After the experimental period, the animals were subjected to the glucose tolerance test (GTT) and plasma non-esterified fatty acids (NEFA) concentration measurements. The soleus skeletal muscle, liver and white adipose tissue were removed for molecular (western blotting and RT-PCR) and histological analysis. RESULTS The beta hydroxy beta methylbutyrate supplemented rats presented: (i) higher ratio between the area under the curve (AUC) of insulinaemia and glycaemia during glucose tolerance test; (ii) impairment of insulin sensitivity on liver and soleus skeletal muscle after insulin overload; (iii) reduction of glucose transporter 4 (GLUT 4) total and plasma membrane content on soleus; (iv) increased hormone-sensitive lipase (HSL) mRNA and protein expression on white adipose tissue and plasma NEFA levels and (v) reduction of fibre cross-sectional area of soleus muscle. CONCLUSION The data altogether indicate that beta hydroxy beta methylbutyrate supplementation impairs insulin sensitivity in healthy sedentary rats, which, in the long-term, could lead to an increased risk of developing type 2 diabetes.
Collapse
Affiliation(s)
- C. Y. Yonamine
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| | - S. S. Teixeira
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| | - R. S. Campello
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| | - F. Gerlinger-Romero
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| | - C. F. Rodrigues
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| | - L. Guimarães-Ferreira
- Department of Sports; Center of Physical Education and Sports; Federal University of Espírito Santo; Vitória Brazil
| | - U. F. Machado
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| | - M. T. Nunes
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| |
Collapse
|
48
|
Borges P, Valente LMP, Véron V, Dias K, Panserat S, Médale F. High dietary lipid level is associated with persistent hyperglycaemia and downregulation of muscle Akt-mTOR pathway in Senegalese sole (Solea senegalensis). PLoS One 2014; 9:e102196. [PMID: 25036091 PMCID: PMC4103825 DOI: 10.1371/journal.pone.0102196] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 06/16/2014] [Indexed: 12/25/2022] Open
Abstract
High levels of dietary lipids are incorporated in feeds for most teleost fish to promote growth and reduce nitrogen waste. However, in Senegalese sole (Solea senegalensis) previous studies revealed that increasing the level of dietary lipids above 8% negatively affect growth and nutrient utilization regardless of dietary protein content. It has been shown that glucose regulation and metabolism can be impaired by high dietary fat intake in mammals, but information in teleost fish is scarce. The aim of this study was to assess the possible effect of dietary lipids on glucose metabolism in Senegalese sole with special emphasis on the regulation of proteins involved in the muscle insulin-signalling pathway. Senegalese sole juveniles (29 g) were fed two isonitrogenous diets (53% dry matter) for 88 days. These two diets were one with a high lipid level (∼17%, HL) and a moderate starch content (∼14%, LC), and the other being devoid of fish oil (4% lipid, LL) and with high starch content (∼23%, HC). Surprisingly, feeding Senegalese sole the HL/LC diet resulted in prolonged hyperglycaemia, while fish fed on LL/HC diet restored basal glycaemia 2 h after feeding. The hyperglycaemic phenotype was associated with greater glucose-6-phosphatase activity (a key enzyme of hepatic glucose production) and lower citrate synthase activity in the liver, with significantly higher liver glycogen content. Sole fed on HL/LC diet also had significantly lower hexokinase activity in muscle, although hexokinase activity was low with both dietary treatments. The HL/LC diet was associated with significant reductions in muscle AKT, p70 ribosomal S6-K1 Kinase (S6K-1) and ribosomal protein S6 (S6) 2 h after feeding, suggesting down regulation of the AKT-mTOR nutrient signalling pathway in these fish. The results of this study show for the first time that high level of dietary lipids strongly affects glucose metabolism in Senegalese sole.
Collapse
Affiliation(s)
- Pedro Borges
- CIMAR/CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental and ICBAS, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- INRA-UR 1067 Nutrition Métabolisme Aquaculture, Pôle Hydrobiologie, Saint Pée-sur-Nivelle, France
| | - Luísa M. P. Valente
- CIMAR/CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental and ICBAS, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Vincent Véron
- INRA-UR 1067 Nutrition Métabolisme Aquaculture, Pôle Hydrobiologie, Saint Pée-sur-Nivelle, France
| | - Karine Dias
- INRA-UR 1067 Nutrition Métabolisme Aquaculture, Pôle Hydrobiologie, Saint Pée-sur-Nivelle, France
| | - Stéphane Panserat
- INRA-UR 1067 Nutrition Métabolisme Aquaculture, Pôle Hydrobiologie, Saint Pée-sur-Nivelle, France
| | - Françoise Médale
- INRA-UR 1067 Nutrition Métabolisme Aquaculture, Pôle Hydrobiologie, Saint Pée-sur-Nivelle, France
- * E-mail:
| |
Collapse
|
49
|
Chakraborty C, Doss CGP, Bandyopadhyay S, Agoramoorthy G. Influence of miRNA in insulin signaling pathway and insulin resistance: micro-molecules with a major role in type-2 diabetes. WILEY INTERDISCIPLINARY REVIEWS-RNA 2014; 5:697-712. [PMID: 24944010 DOI: 10.1002/wrna.1240] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 04/12/2014] [Accepted: 04/17/2014] [Indexed: 12/25/2022]
Abstract
The prevalence of type-2 diabetes (T2D) is increasing significantly throughout the globe since the last decade. This heterogeneous and multifactorial disease, also known as insulin resistance, is caused by the disruption of the insulin signaling pathway. In this review, we discuss the existence of various miRNAs involved in regulating the main protein cascades in the insulin signaling pathway that affect insulin resistance. The influence of miRNAs (miR-7, miR-124a, miR-9, miR-96, miR-15a/b, miR-34a, miR-195, miR-376, miR-103, miR-107, and miR-146) in insulin secretion and beta (β) cell development has been well discussed. Here, we highlight the role of miRNAs in different significant protein cascades within the insulin signaling pathway such as miR-320, miR-383, miR-181b with IGF-1, and its receptor (IGF1R); miR-128a, miR-96, miR-126 with insulin receptor substrate (IRS) proteins; miR-29, miR-384-5p, miR-1 with phosphatidylinositol 3-kinase (PI3K); miR-143, miR-145, miR-29, miR-383, miR-33a/b miR-21 with AKT/protein kinase B (PKB) and miR-133a/b, miR-223, miR-143 with glucose transporter 4 (GLUT4). Insulin resistance, obesity, and hyperlipidemia (high lipid levels in the blood) have a strong connection with T2D and several miRNAs influence these clinical outcomes such as miR-143, miR-103, and miR-107, miR-29a, and miR-27b. We also corroborate from previous evidence how these interactions are related to insulin resistance and T2D. The insights highlighted in this review will provide a better understanding on the impact of miRNA in the insulin signaling pathway and insulin resistance-associated diagnostics and therapeutics for T2D.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Bio-informatics, School of Computer and Information Sciences, Galgotias University, Greater Noida, India
| | | | | | | |
Collapse
|
50
|
Hemer S, Konrad C, Spiliotis M, Koziol U, Schaack D, Förster S, Gelmedin V, Stadelmann B, Dandekar T, Hemphill A, Brehm K. Host insulin stimulates Echinococcus multilocularis insulin signalling pathways and larval development. BMC Biol 2014; 12:5. [PMID: 24468049 PMCID: PMC3923246 DOI: 10.1186/1741-7007-12-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 01/21/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The metacestode of the tapeworm Echinococcus multilocularis is the causative agent of alveolar echinococcosis, a lethal zoonosis. Infections are initiated through establishment of parasite larvae within the intermediate host's liver, where high concentrations of insulin are present, followed by tumour-like growth of the metacestode in host organs. The molecular mechanisms determining the organ tropism of E. multilocularis or the influences of host hormones on parasite proliferation are poorly understood. RESULTS Using in vitro cultivation systems for parasite larvae we show that physiological concentrations (10 nM) of human insulin significantly stimulate the formation of metacestode larvae from parasite stem cells and promote asexual growth of the metacestode. Addition of human insulin to parasite larvae led to increased glucose uptake and enhanced phosphorylation of Echinococcus insulin signalling components, including an insulin receptor-like kinase, EmIR1, for which we demonstrate predominant expression in the parasite's glycogen storage cells. We also characterized a second insulin receptor family member, EmIR2, and demonstrated interaction of its ligand binding domain with human insulin in the yeast two-hybrid system. Addition of an insulin receptor inhibitor resulted in metacestode killing, prevented metacestode development from parasite stem cells, and impaired the activation of insulin signalling pathways through host insulin. CONCLUSIONS Our data indicate that host insulin acts as a stimulant for parasite development within the host liver and that E. multilocularis senses the host hormone through an evolutionarily conserved insulin signalling pathway. Hormonal host-parasite cross-communication, facilitated by the relatively close phylogenetic relationship between E. multilocularis and its mammalian hosts, thus appears to be important in the pathology of alveolar echinococcosis. This contributes to a closer understanding of organ tropism and parasite persistence in larval cestode infections. Furthermore, our data show that Echinococcus insulin signalling pathways are promising targets for the development of novel drugs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Klaus Brehm
- University of Würzburg, Institute of Hygiene and Microbiology, Josef-Schneider-Strasse 2, D-97080 Würzburg, Germany.
| |
Collapse
|