1
|
Zhang X, Jin Y. DUSP9-mediated inhibition of IRS1/PI3K/AKT pathway contributes to insulin resistance and metabolic dysfunction in gestational diabetes mellitus. Hum Immunol 2025; 86:111263. [PMID: 40020430 DOI: 10.1016/j.humimm.2025.111263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/14/2025] [Accepted: 02/10/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is a common pregnancy complication associated with adverse maternal and fetal outcomes. Recent studies suggest a role for dual-specificity phosphatase 9 (DUSP9) in insulin resistance and metabolic dysregulation, though its specific contribution to GDM remains unclear. This study aims to investigate the function of DUSP9 in GDM pathophysiology and its underlying molecular mechanisms. METHODS We analyzed DUSP9 expression in umbilical cord blood and placental tissues from GDM patients (n = 16) and healthy controls (n = 14) using RT-qPCR and western blot assays. In vitro, functional assays were conducted on high glucose-induced HTR-8/SVneo trophoblast cells to evaluate the effects of DUSP9 knockdown on cell viability, apoptosis, and insulin signaling. In vivo, a GDM mouse model was constructed, and lentivirus-mediated shRNA was used to downregulate DUSP9 expression. Furthermore, metabolic parameters, including insulin resistance indices, lipid metabolism, and placental apoptosis were assessed, along with the phosphorylation status of key proteins in the IRS1/PI3K/AKT pathway. RESULTS We first observed that DUSP9 expression was significantly upregulated in the umbilical cord blood and placental tissues of GDM patients compared to healthy controls (p < 0.01). Using high glucose-induced HTR-8/SVneo trophoblast cells to mimic GDM conditions, we found that downregulation of DUSP9 increased cell viability and inhibited apoptosis (p < 0.01). Mechanistically, co-immunoprecipitation and pull-down assays demonstrated that DUSP9 directly interacts with insulin receptor substrate 1 (IRS1) and inhibits HG-mediated IRS1 phosphorylation at Tyr632, impairing downstream insulin signaling (p < 0.01). In vivo, a GDM mouse model revealed elevated DUSP9 expression, along with significant metabolic dysfunction, including insulin resistance and increased placental apoptosis (p < 0.01). Lentivirus-mediated knockdown of DUSP9 in these mice ameliorated insulin resistance, improved lipid metabolism, and reduced placental apoptosis by improving fasting glucose and insulin levels, lipid profiles, and decreased apoptotic markers (p < 0.01). Moreover, DUSP9 knockdown in these mice promoted activation of the IRS1/PI3K/AKT signaling pathway (p < 0.01). CONCLUSIONS DUSP9 contributes to GDM progression by inhibiting the IRS1/PI3K/AKT pathway, leading to insulin resistance and metabolic dysfunction. The knockdown of DUSP9 ameliorates key pathological features of GDM, including insulin resistance, impaired lipid metabolism, and placental apoptosis, suggesting that targeting DUSP9 may represent a potential therapeutic strategy for GDM.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Obstetrics and Gynecology, Nantong Maternal and Child Health Care Hospital, No. 399 Century Avenue, Nantong, Jiangsu 226000, China
| | - Yanqi Jin
- Department of Obstetrics and Gynecology, Nantong Maternal and Child Health Care Hospital, No. 399 Century Avenue, Nantong, Jiangsu 226000, China.
| |
Collapse
|
2
|
Cao T, Zhou Q, Li F, Wang M, Zhang M, Li X, Zhao H, Zhou Y. Dual-specific phosphatases-8: a new target for clinical disease intervention. J Transl Med 2025; 23:485. [PMID: 40301852 PMCID: PMC12042392 DOI: 10.1186/s12967-025-06499-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/13/2025] [Indexed: 05/01/2025] Open
Abstract
Dual-specific phosphatase-8 (DUSP8), identified as the first gene in a genome-wide association study (GWAS), is implicated in cellular oxidative stress, proliferation, apoptosis, and drug resistance through its negative regulation of the dephosphorylation activities of JNK, ERK, and p38 within the MAPK pathway. Recent studies have shown that DUSP8 plays a pivotal role in the progression of several human diseases, notably colorectal cancer, diabetic kidney disease, and breast cancer. This suggests that DUSP8 may represent a novel target for clinical intervention in these diseases. This review first introduces the biological structure and function of DUSP8, with a focus on its relationship with a series of diseases and the regulatory mechanisms involved. Furthermore, we concentrate on unresolved scientific questions in the current research, aiming to establish a new theoretical foundation for the diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Tingping Cao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Quanling Zhou
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Fujun Li
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Mingyue Wang
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Ming Zhang
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Xiaohui Li
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Hailong Zhao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Ya Zhou
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
- Key Laboratory of Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou, 563000, China.
| |
Collapse
|
3
|
Ejiohuo O, Bilska K, Narożna B, Skibińska M, Kapelski P, Dmitrzak-Węglarz M, Szczepankiewicz A, Pawlak J. The implication of ADRA2A and AVPRIB gene variants in the aetiology of stress-related bipolar disorder. J Affect Disord 2025; 368:249-257. [PMID: 39278467 DOI: 10.1016/j.jad.2024.09.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
OBJECTIVE Bipolar disorder is a complex and severe mental illness characterised by manic and depressive episodes that can be triggered and exacerbated by psychosocial, environmental, and biological stressors. Genetic variations are a risk factor for bipolar disorder. However, the identification of the exact gene variants and genotypes remains complex. This study, therefore, aims to identify the potential association between genotypes of analysed single nucleotide polymorphisms and the presence of a stressor in bipolar disorder patients. METHOD We analysed 114 single nucleotide polymorphisms (SNPs) from bipolar and stress-related candidate genes in 550 patients with bipolar disorders (60.36 % females and 39.64 % male). We compared SNPs of patients reporting the presence (40.73 %) or absence of stressors (59.27 %) before the first episode using the Persons Chi-square test and Bayes Factor t-test. The genotyping of 114 SNPs was done using TaqMan assays. Statistical analysis was done using Statistica 13.3 software (StatSoft Poland, Krakow, Poland), R programming, and G*Power statistics. RESULT We found significant differences in genotype distribution (p < 0.05) in 6 polymorphisms (AVPRIB/rs28536160, FKBP4/rs2968909, ADRA2A/rs3750625, 5HTR2A/rs6311, 5HTR2A/rs6313, and GLCCI1/rs37972) when comparing BD patient with and without stressor with a small effect of d = 0.2. Of these, two gene variants (ADRA2A/rs3750625/AC and AVPRIB/rs28536160/CT) with minor alleles formed an association with the presence of a stressor prior to the disease onset and favoured the alternative hypothesis using Bayes Factor Analysis t-test for hypothesis testing. CONCLUSION This study presents a novel association of ADRA2A/rs3750625/AC and AVPR1B/rs28536160/CT gene variants in stress-related bipolar disorder with the AC genotype of ADRA2A/rs3750625 constituting a risk genotype and CT of AVPR1B/rs28536160 constituting a protective genotype. However, further functional analysis is required to fully understand their clinical and biological significance and interaction.
Collapse
Affiliation(s)
- Ovinuchi Ejiohuo
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland; Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland; Molecular and Cell Biology Unit, Poznan University of Medical Sciences, Poznan, Poland.
| | - Karolina Bilska
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Beata Narożna
- Molecular and Cell Biology Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Maria Skibińska
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Paweł Kapelski
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | - Joanna Pawlak
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland.
| |
Collapse
|
4
|
Wang S, Ren Y, Li J, Li H, Li J, Lan X, Wang Y. MicroRNA-671-5p regulates the inflammatory response of periodontal ligament stem cells via the DUSP8/p38 MAPK pathway. Mol Biol Rep 2024; 51:644. [PMID: 38727958 DOI: 10.1007/s11033-024-09510-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/02/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND MicroRNAs are differentially expressed in periodontitis tissues. They are involved in cellular responses to inflammation and can be used as markers for diagnosing periodontitis. Microarray analysis showed that the expression level of microRNA-671-5p in periodontal tissues of patients with periodontitis was increased. In this study, we investigated the mechanism of action of microRNA-671-5p in human periodontal ligament stem cells (hPDLSCs) under inflammatory conditions. METHODS AND RESULTS HPDLSCs were treated with lipopolysaccharide (LPS) to establish an inflammation model. The cell survival rate was determined using the cell counting kit-8 (CCK8). Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot analyses were used to detect the expression of microRNA-671-5p and dual-specificity phosphatase (DUSP) 8 proteins, respectively, Interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α were detected using qRT-PCR and Enzyme-linked immunosorbent assay (ELISA). A dual-luciferase reporter system was employed to determine the relationship between micoRNA-671-5p and DUSP8 expression. Activation of the p38 mitogen-activated protein kinase (MAPK) signaling pathway was confirmed using western blot analysis. Following the treatment of hPDLSCs with LPS, the expression levels of microRNA-671-5p in hPDLSCs were increased, cell viability decreased, and the expression of inflammatory factors displayed an increasing trend. MicroRNA-671-5p targets and binds to DUSP8. Silencing microRNA-671-5p or overexpressing DUSP8 can improve cell survival rate and reduce inflammatory responses. When DUSP8 was overexpressed, the expression of p-p38 was reduced. CONCLUSIONS microRNA-671-5p targets DUSP8/p38 MAPK pathway to regulate LPS-induced proliferation and inflammation in hPDLSCs.
Collapse
Affiliation(s)
- Shifen Wang
- Southwest Medical University, Luzhou, 646000, China
- The Department of Preventive Dentistry, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Yajiao Ren
- Southwest Medical University, Luzhou, 646000, China
- The Department of Preventive Dentistry, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Jiaxin Li
- Southwest Medical University, Luzhou, 646000, China
- The Department of Preventive Dentistry, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Hao Li
- Southwest Medical University, Luzhou, 646000, China
- The Department of Preventive Dentistry, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Jianmin Li
- Southwest Medical University, Luzhou, 646000, China
- The Department of Preventive Dentistry, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Luzhou, 646000, China
| | - Yao Wang
- Southwest Medical University, Luzhou, 646000, China.
- The Department of Preventive Dentistry, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China.
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Luzhou, 646000, China.
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
5
|
Lu J, Ma Y, Lv H, Li C, Ru L, Zhao J, Wang D. The Different Therapeutic Effects of Traditional Chinese Medicine Shensong Yangxin Capsule and Salubrinal in High-intensity Exercise-induced Heart Failure in Rats with Acute Myocardial Infarction. Comb Chem High Throughput Screen 2024; 27:1592-1601. [PMID: 38305401 DOI: 10.2174/0113862073272407231201071629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND Currently, endoplasmic reticulum stress is studied utilizing a dephosphorylation inhibitor (Sal). The traditional Chinese patent medicine and simple formulation Shensong Yangxin Capsule is a commonly used medication for the treatment of arrhythmia. However, the efficacy and underlying mechanism of the capsule in treating post-ischemic heart failure in myocardial tissue have not yet been investigated. OBJECTIVE The therapeutic effects and the underlying mechanism of the Shensong Yangxin Capsule (SSYX) and the dephosphorylation inhibitor Salubrinal (Sal) on heart failure (HF) induced by high-intensity exercise in rats with acute myocardial infarction (AMI) were investigated. METHODS Male infants of 8 weeks Spragge-Dawley (SD) rats were randomly assigned to one of four groups: sham surgery group, AMI+placebo group, AMI+Shensong Yangxin Capsule group (AMI+SSYX), and AMI+Sal administration group. Rats' myocardial infarction was induced by left coronary artery ligation. Rats were subjected to a 3-week high-intensity exercise program to simulate heart failure after 7 days of postoperative rest. After the fourth postoperative week, echocardiography was applied to determine the left ventricular ejection fraction (LVEF), left ventricular fractional shortening (LVFS), and left ventricular systolic volume (LVESV) in each group. HE and TUNEL labeling were employed to examine the morphology of cardiac cells and measure the percentage of apoptosis in each group; Western blotting was applied to detect the cardiomyocyte apoptosis-related proteins p-JNK, p-P38, and NOX2, while ELISA was used to detect glutathione(GSH), malondialdehyde (MDA), and superoxide dismutase (SOD) in serum. RESULTS Following a 4-week drug intervention:(1)LVFS and LVEF in the AMI+placebo group were statistically significantly reduced, while LVESV were significantly higher, compared to those in the sham surgery group (P<0.05); The AMI+SSYX group performed statistically significantly better than the AMI+placebo group(P<0.05). (2) The myocardial cells in the AMI+placebo group exhibited significant swelling and inflammatory cell infiltration; the myocardial cells in the AMI+SSYX group and AMI+Sal group displayed mild swelling and minimal inflammatory cell infiltration; the AMI+SSYX group's myocardial cell morphology was superior to that of the AMI+Sal group; (3) The apoptosis rate of the AMI+placebo group was around 95%, greater than that of the sham surgery group (2.55%). The apoptosis rate of the AMI+SSYX group is approximately 21%, while the apoptosis rate of the AMI+Sal group is about 43%. (4) In the AMI+placebo group, p-JNK, p-P38, and NOX2 protein expression dramatically increased compared to the sham surgery group. The expression of p-P38, NOX2, and p-JNK/t-JNK was considerably reduced in the AMI+Shensong group and AMI+Sal group, compared to the AMI+placebo group. (P<0.01)The AMI+SSYX group's result is superior to that of the AMI+Sal group. (5) Compared to the sham surgery group, the serum levels of SOD and GSH were significantly lower, and MDA was significantly higher in the AMI+placebo group. Compared to the AMI+placebo group, the serum levels of SOD and GSH were significantly higher, and MDA was significantly lower in the AMI+SSYX group and the AMI+Sal group. (P<0.05) Conclusion: In rats with acute myocardial infarction in high-intensity exercise-induced heart failure, Shensong Yangxin Capsule dramatically reduces myocardial cell death and cardiac dysfunction. SSYX has a shorter course of treatment and a better therapeutic effect than Sal.
Collapse
Affiliation(s)
- Junli Lu
- Department of Radiology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yanzhuo Ma
- Internal Medicine Teaching and Research Section, Hebei Medical University; No.361 Zhongshan Road, Shijiazhuang, 050017, China
| | - Hongzhi Lv
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Congxin Li
- Department of Pharmacy, The Third Hospital of Hebei Medical University, Shijiazhuang, 050001, China
| | - Leisheng Ru
- Internal Medicine Teaching and Research Section, Hebei Medical University; No.361 Zhongshan Road, Shijiazhuang, 050017, China
| | - Jian Zhao
- Department of Radiology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Dongmei Wang
- Internal Medicine Teaching and Research Section, Hebei Medical University; No.361 Zhongshan Road, Shijiazhuang, 050017, China
- Department of Cardiology, 980 Hospital of PLA Joint Logistic Support Forces, Shijiazhuang, 050000, China
| |
Collapse
|
6
|
Chuang HC, Hsueh CH, Hsu PM, Tsai CY, Shih YC, Chiu HY, Chen YM, Yu WK, Chen MH, Tan TH. DUSP8 induces TGF-β-stimulated IL-9 transcription and Th9-mediated allergic inflammation by promoting nuclear export of Pur-α. J Clin Invest 2023; 133:e166269. [PMID: 37909329 PMCID: PMC10617771 DOI: 10.1172/jci166269] [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: 10/13/2022] [Accepted: 09/07/2023] [Indexed: 11/03/2023] Open
Abstract
Dual-specificity phosphatase 8 (DUSP8) is a MAPK phosphatase that dephosphorylates and inactivates the kinase JNK. DUSP8 is highly expressed in T cells; however, the in vivo role of DUSP8 in T cells remains unclear. Using T cell-specific Dusp8 conditional KO (T-Dusp8 cKO) mice, mass spectrometry analysis, ChIP-Seq, and immune analysis, we found that DUSP8 interacted with Pur-α, stimulated interleukin-9 (IL-9) gene expression, and promoted Th9 differentiation. Mechanistically, DUSP8 dephosphorylated the transcriptional repressor Pur-α upon TGF-β signaling, leading to the nuclear export of Pur-α and subsequent IL-9 transcriptional activation. Furthermore, Il-9 mRNA levels were induced in Pur-α-deficient T cells. In addition, T-Dusp8-cKO mice displayed reduction of IL-9 and Th9-mediated immune responses in the allergic asthma model. Reduction of Il-9 mRNA levels in T cells and allergic responses of T-Dusp8-cKO mice was reversed by Pur-α knockout. Remarkably, DUSP8 protein levels and the DUSP8-Pur-α interaction were indeed increased in the cytoplasm of T cells from people with asthma and patients with atopic dermatitis. Collectively, DUSP8 induces TGF-β-stimulated IL-9 transcription and Th9-induced allergic responses by inhibiting the nuclear translocation of the transcriptional repressor Pur-α. DUSP8 may be a T-cell biomarker and therapeutic target for asthma and atopic dermatitis.
Collapse
Affiliation(s)
- Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Chia-Hsin Hsueh
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Pu-Ming Hsu
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Ching-Yi Tsai
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Ying-Chun Shih
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Hsien-Yi Chiu
- Department of Dermatology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Yi-Ming Chen
- Division of Allergy, Immunology, and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | | | - Ming-Han Chen
- Division of Allergy, Immunology, and Rheumatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
7
|
Munyaneza JP, Kim M, Cho E, Jang A, Choo HJ, Lee JH. Association of single-nucleotide polymorphisms in dual specificity phosphatase 8 and insulin-like growth factor 2 genes with inosine-5'-monophosphate, inosine, and hypoxanthine contents in chickens. Anim Biosci 2023; 36:1357-1366. [PMID: 37402464 PMCID: PMC10472161 DOI: 10.5713/ab.23.0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/21/2023] [Accepted: 05/22/2023] [Indexed: 07/06/2023] Open
Abstract
OBJECTIVE This study aimed to identify the single-nucleotide polymorphisms (SNPs) in the dual-specificity phosphatase 8 (DUSP8) and insulin-like growth factor 2 (IGF2) genes and to explore their effects on inosine-5'-monophosphate (IMP), inosine, and hypoxanthine contents in Korean native chicken -red-brown line (KNC-R Line). METHODS A total sample of 284 (males, n = 127; females n = 157) and 230 (males, n = 106; females, n = 124) aged of 10 weeks old KNC-R line was used for genotyping of DUSP8 and IGF2 genes, respectively. One SNP (rs313443014 C>T) in DUSP8 gene and two SNPs (rs315806609A/G and rs313810945T/C) in IGF2 gene were used for genotyping by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) and KASP methods, respectively. The Two-way analysis of variance of the R program was used to associate DUSP8 and IGF2 genotypes with nucleotide contents in KNC-R chickens. RESULTS The DUSP8 (rs313443014 C>T) was polymorphic in KNC-R line and showed three genotypes: CC, CT, and TT. The IGF2 gene (rs315806609A/G and rs313810945T/C) was also polymorphic and had three genotypes per SNP, including GG, AG, and AA for the SNP rs315806609A/G and genotypes: CC, CT, and TT for the SNP rs313810945T/C. Association resulted into a strong significant association (p<0.01) with IMP, inosine, and hypoxanthine. Moreover, the significant effect of sex (p<0.05) on nucleotide content was also observed. CONCLUSION The SNPs in the DUSP8 and IGF2 genes might be used as genetic markers in the selection and production of chickens with highly flavored meat.
Collapse
Affiliation(s)
- Jean Pierre Munyaneza
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134,
Korea
| | - Minjun Kim
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134,
Korea
| | - Eunjin Cho
- Department of Bio-AI Convergence, Chungnam National University, Daejeon 34134,
Korea
| | - Aera Jang
- Department of Applied Animal Science, College of Animal Life Science, Kangwon National University, Chuncheon 24341,
Korea
| | - Hyo Jun Choo
- Poultry Research Institute, National Institute of Animal Science, Pyeongchang 25342,
Korea
| | - Jun Heon Lee
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134,
Korea
- Department of Bio-AI Convergence, Chungnam National University, Daejeon 34134,
Korea
| |
Collapse
|
8
|
Huang W, Lao L, Deng Y, Li Z, Liao W, Duan S, Xiao S, Cao Y, Miao J. Preparation, characterization, and osteogenic activity mechanism of casein phosphopeptide-calcium chelate. Front Nutr 2022; 9:960228. [PMID: 35983483 PMCID: PMC9378869 DOI: 10.3389/fnut.2022.960228] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/07/2022] [Indexed: 12/25/2022] Open
Abstract
Casein phosphopeptides (CPPs) are good at calcium-binding and intestinal calcium absorption, but there are few studies on the osteogenic activity of CPPs. In this study, the preparation of casein phosphopeptide calcium chelate (CPP-Ca) was optimized on the basis of previous studies, and its peptide-calcium chelating activity was characterized. Subsequently, the effects of CPP-Ca on the proliferation, differentiation, and mineralization of MC3T3-E1 cells were studied, and the differentiation mechanism of CPP-Ca on MC3T3-E1 cells was further elucidated by RNA sequencing (RNA-seq). The results showed that the calcium chelation rate of CPPs was 23.37%, and the calcium content of CPP-Ca reached 2.64 × 105 mg/kg. The test results of Ultraviolet–Visible absorption spectroscopy (UV) and Fourier transform infrared spectroscopy (FTIR) indicated that carboxyl oxygen and amino nitrogen atoms of CPPs might be chelated with calcium during the chelation. Compared with the control group, the proliferation of MC3T3-E1 cells treated with 250 μg/mL of CPP-Ca increased by 21.65%, 26.43%, and 28.43% at 24, 48, and 72 h, respectively, and the alkaline phosphatase (ALP) activity and mineralized calcium nodules of MC3T3-E1 cells were notably increased by 55% and 72%. RNA-seq results showed that 321 differentially expressed genes (DEGs) were found in MC3T3-E1 cells treated with CPP-Ca, including 121 upregulated and 200 downregulated genes. Gene ontology (GO) revealed that the DEGs mainly played important roles in the regulation of cellular components. The enrichment of the Kyoto Encyclopedia of Genes and Genomes Database (KEGG) pathway indicated that the AMPK, PI3K-Akt, MAPK, and Wnt signaling pathways were involved in the differentiation of MC3T3-E1 cells. The results of a quantitative real-time PCR (qRT-PCR) showed that compared with the blank control group, the mRNA expressions of Apolipoprotein D (APOD), Osteoglycin (OGN), and Insulin-like growth factor (IGF1) were significantly increased by 2.6, 2.0 and 3.0 times, respectively, while the mRNA levels of NOTUM, WIF1, and LRP4 notably decreased to 2.3, 2.1, and 4.2 times, respectively, which were consistent both in GO functional and KEGG enrichment pathway analysis. This study provided a theoretical basis for CPP-Ca as a nutritional additive in the treatment and prevention of osteoporosis.
Collapse
Affiliation(s)
- Wen Huang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Linhui Lao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Yuliang Deng
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Ziwei Li
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Wanwen Liao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Shan Duan
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Suyao Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Jianyin Miao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Guangxi Normal University), Guilin, China.,Solid-State Fermentation Resource Utilization Key Laboratory of Sichuan Province, Yibin, China
| |
Collapse
|
9
|
Pujol-Gualdo N, Läll K, Lepamets M, Rossi HR, Arffman RK, Piltonen TT, Mägi R, Laisk T. Advancing our understanding of genetic risk factors and potential personalized strategies for pelvic organ prolapse. Nat Commun 2022; 13:3584. [PMID: 35739095 PMCID: PMC9226158 DOI: 10.1038/s41467-022-31188-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 06/08/2022] [Indexed: 11/09/2022] Open
Abstract
Pelvic organ prolapse is a common gynecological condition with limited understanding of its genetic background. In this work, we perform a genome-wide association meta-analysis comprising 28,086 cases and 546,291 controls from European ancestry. We identify 19 novel genome-wide significant loci, highlighting connective tissue, urogenital and cardiometabolic as likely affected systems. Here, we prioritize many genes of potential interest and assess shared genetic and phenotypic links. Additionally, we present the first polygenic risk score, which shows similar predictive ability (Harrell C-statistic (C-stat) 0.583, standard deviation (sd) = 0.007) as five established clinical risk factors combined (number of children, body mass index, ever smoked, constipation and asthma) (C-stat = 0.588, sd = 0.007) and demonstrates a substantial incremental value in combination with these (C-stat = 0.630, sd = 0.007). These findings improve our understanding of genetic factors underlying pelvic organ prolapse and provide a solid start evaluating polygenic risk scores as a potential tool to enhance individual risk prediction. Although pelvic organ prolapse is a common gynecological condition, the genetic component of disease risk is not well known. Here the authors find common genetic variants associated with the disease and present a polygenic risk score to enhance individual risk prediction.
Collapse
Affiliation(s)
- Natàlia Pujol-Gualdo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia. .,Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu, University Hospital, University of Oulu, Oulu, Finland.
| | - Kristi Läll
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Maarja Lepamets
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | | | - Henna-Riikka Rossi
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu, University Hospital, University of Oulu, Oulu, Finland
| | - Riikka K Arffman
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu, University Hospital, University of Oulu, Oulu, Finland
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu, University Hospital, University of Oulu, Oulu, Finland
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| |
Collapse
|
10
|
Wang J, Zhou CC, Sun HC, Li Q, Hu JD, Jiang T, Zhou S. Identification of several senescence-associated genes signature in head and neck squamous cell carcinoma. J Clin Lab Anal 2022; 36:e24555. [PMID: 35692082 PMCID: PMC9279997 DOI: 10.1002/jcla.24555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022] Open
Abstract
Background As one of the core aging processes, cellular senescence is associated with tumorigenesis, growth, and immune modulation in cancers. Nevertheless, the prognosis of senescence‐associated genes (SAGs) signature in head and neck squamous cell carcinoma (HNSCC) remains to be further evaluated. Methods The transcriptome and corresponding clinical datasets of SAGs in patients with HNSCC were downloaded from public databases. A new prognostic SAGs signature was established with least absolute shrinkage and selection operator discussion. Patients with HNSCC were fallen into two risk groups based on each sample's risk mark and the cutoff point. The survival analysis was extended to determine the predictive accuracy of the SAGs signature. Furthermore, the evaluation of SAGs signature was made according to clinicopathological characteristics, survival state, the infiltration of inflammatory cells, and efficacy of immunotherapy. Results 41 SAGs were recognized and adopted to establish the forecast signature. The survival analysis indicated that patients with HNSCC in the high‐senescent score group had significantly reduced overall survival compared with those in the low‐senescent score group. It was certified that the risk score of SAGs signature was a separate predicting agent for HNSCC applying Cox regression analysis. According to functional analysis, some immune‐associated pathways were increased in the low‐senescent score group significantly. High‐senescent score group was correlated with poor clinicopathological characteristics, given less the infiltration of inflammatory cells state and worse immunotherapeutic effect. Conclusion A new SAG signature predicting result and response to immunotherapy of HNSCC was identified. Cellular senescence may be a hidden target for HNSCC.
Collapse
Affiliation(s)
- Jian Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Chong-Chang Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Hong-Cun Sun
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Qun Li
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Jian-Dao Hu
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Tao Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Shao Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
11
|
Díaz-Chamorro S, Garrido-Jiménez S, Barrera-López JF, Mateos-Quirós CM, Cumplido-Laso G, Lorenzo MJ, Román ÁC, Bernardo E, Sabio G, Carvajal-González JM, Centeno F. Title: p38δ Regulates IL6 Expression Modulating ERK Phosphorylation in Preadipocytes. Front Cell Dev Biol 2022; 9:708844. [PMID: 35111744 PMCID: PMC8802314 DOI: 10.3389/fcell.2021.708844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022] Open
Abstract
IL6 is an essential cytokine in metabolism regulation and for intercommunication among different organs and tissues. IL6 produced by different tissues has different functions and therefore it is very important to understand the mechanism of its expression in adipose tissue. In this work we demonstrated that IL6 expression in mouse preadipocytes, like in human, is partially dependent on Wnt5a and JNK. Using mouse preadipocytes lacking each one of the p38 SAPK family members, we have shown that IL6 expression is also p38γ and p38δ dependent. In fact, the lack of some of these two kinases increases IL6 expression without altering that of Wnt5a. Moreover, we show that the absence of p38δ promotes greater ERK1/2 phosphorylation in a MEK1/2 independent manner, and that this increased ERK1/2 phosphorylation state is contributing to the higher IL6 expression in p38δ−/- preadipocytes. These results suggest a new crosstalk between two MAPK signaling pathway, p38δ and ERK1/2, where p38δ modulates the phosphorylation state of ERK1/2.
Collapse
Affiliation(s)
- Selene Díaz-Chamorro
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Sergio Garrido-Jiménez
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Juan Francisco Barrera-López
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Clara María Mateos-Quirós
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Guadalupe Cumplido-Laso
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - María Jesús Lorenzo
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Cáceres, Spain
| | - Ángel Carlos Román
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Edgar Bernardo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - José María Carvajal-González
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Francisco Centeno
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| |
Collapse
|
12
|
Shi J, Li R, Yang Y, Ji L, Li C. Protective effect of α-asarone and β-asarone on Aβ -induced inflammatory response in PC12 cells and its. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 50:591-600. [PMID: 34986541 PMCID: PMC8732252 DOI: 10.3724/zdxbyxb-2021-0162] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/18/2021] [Indexed: 11/25/2022]
Abstract
To investigate effects of α-asarone and β-asarone on induced PC12 cell injury and related mechanisms. Aβ toxic injury cell model was induced by Aβ in PC12 cells. PC12 cells were divided into blank control group, model control group, α-asarone group (0.5, 1.0, β-asarone group (6.3, 12.5, vasoactive intestinal peptide (VIP) group, and VIP antagonist control group. Cell survival rate was detected by CCK-8 kit; cell apoptosis rate was detected by flow cytometry. The levels of inflammatory cytokines interleukin (IL)-1, , tumor necrosis factor (TNF)-α, oxidation-related inducible nitric oxide synthase (iNOS), nitric oxide (NO), apoptosis factors caspase-3 and p53 were detected by ELISA method. The expressions of C-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38MAPK) were detected by Western blotting. Compared with model control group, cell survival rates of group, β-asarone group and VIP group increased; the cell apoptosis rate decreased; levels of apoptosis-related factors caspase-3, p53, inflammatory factors IL-1, TNF-α decreased; IL-10 level increased; levels of oxidization-related factors iNOS and NO decreased; the expression of JNK and p38MAPK protein decreased (all <0.05). After VIP antagonist intervention, the survival rate of β-asarone group decreased; apoptosis rate increased; apoptosis related factors caspase-3, p53, inflammatory factors IL-1, TNF-α increased; IL-10 decreased; oxidation related factors iNOS and NO increased; the expression of JNK and p38MAPK protein increased (all <0.05); while there were no significant changes in these indicators of α-asarone group (all >0.05). α-asarone and β-asarone have protective effects on PC12 cell injury induced by Aβ. β-asarone may inhibit inflammatory factors and oxidation-related factors through promoting VIP secretion, regulating JNK/MAPK pathway, and reducing PC12 cell apoptosis; however, the effect of α-asarone may be not related to VIP secretion.
Collapse
Affiliation(s)
- Jianhong Shi
- 3. School of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China
| | - Ruizhi Li
- 3. School of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China
| | - Yuanxiao Yang
- 3. School of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China
| | - Liting Ji
- 3. School of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China
| | - Changyu Li
- 3. School of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China
| |
Collapse
|
13
|
Schriever SC, Kabra DG, Pfuhlmann K, Baumann P, Baumgart EV, Nagler J, Seebacher F, Harrison L, Irmler M, Kullmann S, Corrêa-da-Silva F, Giesert F, Jain R, Schug H, Castel J, Martinez S, Wu M, Häring HU, de Angelis MH, Beckers J, Müller TD, Stemmer K, Wurst W, Rozman J, Nogueiras R, De Angelis M, Molkentin JD, Krahmer N, Yi CX, Schmidt MV, Luquet S, Heni M, Tschöp MH, Pfluger PT. Type 2 diabetes risk gene Dusp8 regulates hypothalamic Jnk signaling and insulin sensitivity. J Clin Invest 2021; 130:6093-6108. [PMID: 32780722 DOI: 10.1172/jci136363] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022] Open
Abstract
Recent genome-wide association studies (GWAS) identified DUSP8, encoding a dual-specificity phosphatase targeting mitogen-activated protein kinases, as a type 2 diabetes (T2D) risk gene. Here, we reveal that Dusp8 is a gatekeeper in the hypothalamic control of glucose homeostasis in mice and humans. Male, but not female, Dusp8 loss-of-function mice, either with global or corticotropin-releasing hormone neuron-specific deletion, had impaired systemic glucose tolerance and insulin sensitivity when exposed to high-fat diet (HFD). Mechanistically, we found impaired hypothalamic-pituitary-adrenal axis feedback, blunted sympathetic responsiveness, and chronically elevated corticosterone levels driven by hypothalamic hyperactivation of Jnk signaling. Accordingly, global Jnk1 ablation, AAV-mediated Dusp8 overexpression in the mediobasal hypothalamus, or metyrapone-induced chemical adrenalectomy rescued the impaired glucose homeostasis of obese male Dusp8-KO mice, respectively. The sex-specific role of murine Dusp8 in governing hypothalamic Jnk signaling, insulin sensitivity, and systemic glucose tolerance was consistent with functional MRI data in human volunteers that revealed an association of the DUSP8 rs2334499 risk variant with hypothalamic insulin resistance in men. Further, expression of DUSP8 was increased in the infundibular nucleus of T2D humans. In summary, our findings suggest the GWAS-identified gene Dusp8 as a novel hypothalamic factor that plays a functional role in the etiology of T2D.
Collapse
Affiliation(s)
- Sonja C Schriever
- Research Unit NeuroBiology of Diabetes and.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Dhiraj G Kabra
- Research Unit NeuroBiology of Diabetes and.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Biological Research Pharmacology Department, Sun Pharma Advanced Research Company Ltd., Vadodara, India
| | - Katrin Pfuhlmann
- Research Unit NeuroBiology of Diabetes and.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Division of Metabolic Diseases and
| | - Peter Baumann
- Research Unit NeuroBiology of Diabetes and.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Neurobiology of Diabetes, School of Medicine, Technical University of Munich, Munich, Germany
| | - Emily V Baumgart
- Research Unit NeuroBiology of Diabetes and.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | | | - Fabian Seebacher
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Division of Metabolic Diseases and
| | - Luke Harrison
- Research Unit NeuroBiology of Diabetes and.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Division of Metabolic Diseases and
| | - Martin Irmler
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Stephanie Kullmann
- German Center for Diabetes Research (DZD), Neuherberg, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen, Tübingen, Germany
| | - Felipe Corrêa-da-Silva
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Florian Giesert
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Department of Developmental Genetics, School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Ruchi Jain
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital Malmö, Malmö, Sweden
| | - Hannah Schug
- SYNLAB Analytics and Services, Switzerland AG, Dielsdorf, Switzerland
| | - Julien Castel
- Université de Paris, BFA, UMR 8251, CNRS, Paris, France
| | | | - Moya Wu
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD), Neuherberg, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen, Tübingen, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany
| | - Martin Hrabe de Angelis
- German Center for Diabetes Research (DZD), Neuherberg, Germany.,Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany.,Chair of Experimental Genetics, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Johannes Beckers
- German Center for Diabetes Research (DZD), Neuherberg, Germany.,Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany.,Chair of Experimental Genetics, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Pharmacology and Experimental Therapy, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany
| | - Kerstin Stemmer
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Department of Developmental Genetics, School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jan Rozman
- German Center for Diabetes Research (DZD), Neuherberg, Germany.,Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany.,Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Ruben Nogueiras
- Department of Physiology, Instituto de Investigación Sanitaria, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA.,Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Natalie Krahmer
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Mathias V Schmidt
- Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Serge Luquet
- Université de Paris, BFA, UMR 8251, CNRS, Paris, France
| | - Martin Heni
- German Center for Diabetes Research (DZD), Neuherberg, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen, Tübingen, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany.,Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Division of Metabolic Diseases and
| | - Paul T Pfluger
- Research Unit NeuroBiology of Diabetes and.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Neurobiology of Diabetes, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
14
|
Nuclear P38: Roles in Physiological and Pathological Processes and Regulation of Nuclear Translocation. Int J Mol Sci 2020; 21:ijms21176102. [PMID: 32847129 PMCID: PMC7504396 DOI: 10.3390/ijms21176102] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023] Open
Abstract
The p38 mitogen-activated protein kinase (p38MAPK, termed here p38) cascade is a central signaling pathway that transmits stress and other signals to various intracellular targets in the cytoplasm and nucleus. More than 150 substrates of p38α/β have been identified, and this number is likely to increase. The phosphorylation of these substrates initiates or regulates a large number of cellular processes including transcription, translation, RNA processing and cell cycle progression, as well as degradation and the nuclear translocation of various proteins. Being such a central signaling cascade, its dysregulation is associated with many pathologies, particularly inflammation and cancer. One of the hallmarks of p38α/β signaling is its stimulated nuclear translocation, which occurs shortly after extracellular stimulation. Although p38α/β do not contain nuclear localization or nuclear export signals, they rapidly and robustly translocate to the nucleus, and they are exported back to the cytoplasm within minutes to hours. Here, we describe the physiological and pathological roles of p38α/β phosphorylation, concentrating mainly on the ill-reviewed regulation of p38α/β substrate degradation and nuclear translocation. In addition, we provide information on the p38α/β ’s substrates, concentrating mainly on the nuclear targets and their role in p38α/β functions. Finally, we also provide information on the mechanisms of nuclear p38α/β translocation and its use as a therapeutic target for p38α/β-dependent diseases.
Collapse
|
15
|
Histone deacetylase inhibition by MS-275 potentiates glucose-stimulated insulin secretion without affecting glucose oxidation. Life Sci 2020; 257:118073. [PMID: 32663574 DOI: 10.1016/j.lfs.2020.118073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/25/2022]
Abstract
AIMS The preservation of pancreatic beta-cell function is crucial for the treatment of type 2 diabetes. Inhibition of class I histone deacetylase (HDAC) has been proved to protect beta-cells from palmitate- or cytokine-induced apoptosis and increase insulin secretion. However, the underlying molecular mechanism is unclear. MAIN METHODS Rat islets were isolated for insulin secretion, real-time PCR, RNA- sequencing, ChIP-PCR, and oxygen consumption rate analysis after treated with the HDAC1 and HDAC3 inhibitor MS-275. KEY FINDINGS MS-275 pretreatment significantly potentiated insulin secretion from rat islets. RNA-sequencing revealed that multiple signaling pathways were involved in MS-275-regulated islet function. Cacna1g and Adcy1 in calcium and cAMP signaling pathways were up-regulated in MS-275-treated islets, which was validated by real-time PCR. The expressions of the two genes displayed a similar increase in islets isolated from mice treated with MS-275. Knockdown of HDAC1 elevated Cacna1g and Adcy1 expressions in islets. ChIP-sequencing analysis showed that the pan-HDAC inhibitor sodium butyrate increased H3K27 acetylation level in the upstream region of Adcy1 and the promoter region of Cacna1g. ChIP-PCR revealed a similar result in MS-275-treated rat islets. However, MS-275 had minor effect on glucose-induced oxygen consumption rate in rat islets. Unlike glucose, MS-275 did not alter the expressions of glucose-sensitive genes such as Glut2 and Gck, but elevated intracellular Ca2+ concentration in beta-cells. SIGNIFICANCE Our findings support the notion that MS-275-potentiated insulin secretion is involved in calcium and cAMP signaling-mediated gene expressions independent of glucose oxidation. Therefore, HDAC inhibition may serve as a therapeutic strategy for type 2 diabetes.
Collapse
|
16
|
Dong N, Xu X, Xue C, Wang C, Li X, Bi C, Shan A. Ethyl pyruvate inhibits LPS induced IPEC-J2 inflammation and apoptosis through p38 and ERK1/2 pathways. Cell Cycle 2019; 18:2614-2628. [PMID: 31475609 PMCID: PMC6773235 DOI: 10.1080/15384101.2019.1653106] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 07/30/2019] [Accepted: 08/02/2019] [Indexed: 02/07/2023] Open
Abstract
The endotoxin of Gram-negative bacteria threatens the intestinal health of livestock. Ethyl pyruvate (EP) has been shown to regulate intestinal immunity and protect against cell and tissue damage. In this study, it was first verified that EP could reduce the secretion of IL-8, TNF-α, IL-6 and IL-1β in LPS-induced IPEC-J2 cells. Then, we used RNA sequencing (RNA-seq) to analyze the differentially expressed genes (DEGs) of inflammatory factors induced by LPS in IPEC-J2 cells. It was found that LPS induced the upregulation of 377 genes and the downregulation of 477 genes compared to Vehicle; LPS+EP induced the upregulation of 258 genes and the downregulation of 240 genes compared to Vehicle; and LPS+EP induced the upregulation of 373 genes and the downregulation of 188 genes compared to LPS (fold change > 1.5 and FDR < 0.01). Their enrichment pathways included the MAPK signaling pathway, PI3K-Akt signaling pathway, Toll-like receptor signaling pathway, and other pathways. Furthermore, the mRNA level of cytokines associated with inflammation and apoptosis enriched in the MAPK pathway was verified by qRT-PCR. Western blots and immunofluorescence revealed that EP significantly inhibited phosphorylated p38 and phosphorylated-ERK1/2 protein expression levels (P < 0.05). The apoptosis due to LPS reduced by EP was significantly inhibited, as shown by Annexin V-FITC/PI staining. According to the results, EP inhibited the expression of IL-8, TNF-α, IL-6 and IL-1β as well as apoptosis by inhibiting the phosphorylation of p38 and ERK1/2 in LPS-induced IPEC-J2 cells.
Collapse
Affiliation(s)
- Na Dong
- From the Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Xinyao Xu
- From the Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Chenyu Xue
- From the Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Chensi Wang
- From the Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Xinran Li
- From the Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Chongpeng Bi
- From the Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Anshan Shan
- From the Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| |
Collapse
|
17
|
Wu G, Tan J, Li J, Sun X, Du L, Tao S. miRNA-145-5p induces apoptosis after ischemia-reperfusion by targeting dual specificity phosphatase 6. J Cell Physiol 2019; 234:16281-16289. [PMID: 30883744 DOI: 10.1002/jcp.28291] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/04/2018] [Accepted: 12/07/2018] [Indexed: 01/24/2023]
Abstract
Disorders mainly caused by ischemia-reperfusion (I/R), including stroke and myocardial infarction, is linked to debilitating health conditions and death. Recent research indicates that microRNAs (miRNAs) mediate the process of ischemic pathology. This study investigated the effects of miR-145-5p in regulating myocardial ischemic injury. The I/R models were established in rat cardiomyocytes H9C2 and rats. Western blot analysis and quantitative polymerase chain reaction was performed to analyze protein expression. Annexin V-FITC/PI staining was conducted to evaluate cell apoptosis. The application of miR-145-5p mimics and inhibitor revealed that miR-145-5p promoted apoptosis in cardiomyocytes. Furthermore, we found that miR-145-5p directly inhibited dual specificity phosphatase 6 (DUSP6) by luciferase reporter assay. The results indicated that DUSP6 was beneficial against I/R injury through inhibiting c-Jun N-terminal kinase pathways. In conclusion, the essential roles of miR-145-5p and DUSP6 in I/R provide a novel therapeutic target to develop future intervention strategies.
Collapse
Affiliation(s)
- Gang Wu
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jiaying Tan
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Junping Li
- Department of Obstetrics and Gynecology, Huashan Hospital North, Fudan University, Shanghai, People's Republic of China
| | - Xiaoli Sun
- Department of Cardiology, Karamay Central Hospital, Karamay, People's Republic of China
| | - Lei Du
- Department of Cardiology, Karamay Central Hospital, Karamay, People's Republic of China
| | - Sun Tao
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
18
|
Ding T, Zhou Y, Long R, Chen C, Zhao J, Cui P, Guo M, Liang G, Xu L. DUSP8 phosphatase: structure, functions, expression regulation and the role in human diseases. Cell Biosci 2019; 9:70. [PMID: 31467668 PMCID: PMC6712826 DOI: 10.1186/s13578-019-0329-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/12/2019] [Indexed: 12/28/2022] Open
Abstract
Dual-specificity phosphatases (DUSPs) are a subset of protein tyrosine phosphatases (PTPs), many of which dephosphorylate the residues of phosphor-serine/threonine and phosphor-tyrosine on mitogen-activated protein kinases (MAPKs), and hence are also referred to as MAPK phosphatases (MKPs). Homologue of Vaccinia virus H1 phosphatase gene clone 5 (HVH-5), also known as DUSP8, is a unique member of the DUSPs family of phosphatases. Accumulating evidence has shown that DUSP8 plays an important role in phosphorylation-mediated signal transduction of MAPK signaling ranging from cell oxidative stress response, cell apoptosis and various human diseases. It is generally believed that DUSP8 exhibits significant dephosphorylation activity against JNK, however, with the deepening of research, plenty of new literature reports that DUSP8 also has effective dephosphorylation activity on p38 MAPK and ERKs, successfully affects the transduction of MAPKs pathway, indicating that DUSP8 presents a unknown diversity of DUSPs family on distinct corresponding dephosphorylated substrates in different biological events. Therefore, the in-depth study of DUSP8 not only throws a new light on the multi-biological function of DUSPs, but also is much valuable for the reveal of complex pathobiology of clinical diseases. In this review, we provide a detail overview of DUSP8 phosphatase structure, biological function and expression regulation, as well as its role in related clinical human diseases, which might be help for the understanding of biological function of DUSP8 and the development of prevention, diagnosis and therapeutics in related human diseases.
Collapse
Affiliation(s)
- Tao Ding
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China.,2Department of Immunology, Zunyi Medical University, Zunyi, 563000 Guizhou China
| | - Ya Zhou
- 3Department of Medical Physics, Zunyi Medical University, Zunyi, 563000 Guizhou China
| | - Runying Long
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China.,2Department of Immunology, Zunyi Medical University, Zunyi, 563000 Guizhou China
| | - Chao Chen
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China
| | - Panpan Cui
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China
| | - Guiyou Liang
- 4Department of Cardiovascular Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China.,5Department of Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 Guizhou China
| | - Lin Xu
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China.,2Department of Immunology, Zunyi Medical University, Zunyi, 563000 Guizhou China
| |
Collapse
|