1
|
Wang Y, Guo A, Yang L, Han X, Li Q, Liu J, Han Y, Yang Y, Chao L. Immune dysregulation of decidual NK cells mediated by GRIM19 downregulation contributes to the occurrence of recurrent pregnancy loss. Mol Cell Biochem 2024:10.1007/s11010-024-05181-z. [PMID: 39663335 DOI: 10.1007/s11010-024-05181-z] [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: 07/18/2024] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
In patients with recurrent pregnancy loss (RPL), excessive activation of decidual natural killer (dNK) cells has been widely observed, yet the precise underlying mechanisms remain to be elucidated. We collected decidual specimens from RPL patients and controls to assess GRIM19 expression, activation phenotype, cytotoxic function, inflammatory cytokine secretion, and mitochondrial homeostasis in dNK cells. Furthermore, we established a GRIM19-knockout NK-92MI cell line and a GRIM19 ± C57BL/6J mouse model to investigate the relationship between GRIM19 downregulation and dNK immune dysregulation, ultimately contributing to pregnancy loss. Decidual NK cells from RPL patients exhibited significantly lower GRIM19 expression, accompanied by abnormal hyperactivation, enhanced cytotoxicity, and abnormal mitochondrial activation. In vitro experiments confirmed that reduced GRIM19 expression significantly potentiated the cytotoxicity and pro-inflammatory cytokine secretion of NK-92MI cells, while also promoting mitochondrial homeostasis imbalance. Mouse model studies corroborated that GRIM19 downregulation triggers NK cell homeostasis imbalance, contributing to the occurrence of pregnancy loss. Downregulation of GRIM19 in dNK cells contributes to RPL through hyperactivation and disruption of mitochondrial homeostasis, emphasizing its potential as a diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Ying Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Anliang Guo
- Shandong University, Jinan, 250012, Shandong, China
| | - Lin Yang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Xiaojuan Han
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Qianni Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Jin Liu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Yilong Han
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Yang Yang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Lan Chao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
2
|
Gao G, Miao J, Jia Y, He A. Mitochondria-associated programmed cell death: elucidating prognostic biomarkers, immune checkpoints, and therapeutic avenues in multiple myeloma. Front Immunol 2024; 15:1448764. [PMID: 39726602 PMCID: PMC11670199 DOI: 10.3389/fimmu.2024.1448764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background Multiple myeloma (MM) is a hematological malignancy characterized by the abnormal proliferation of plasma cells. Mitochondrial dysfunction and dysregulated programmed cell death (PCD) pathways have been implicated in MM pathogenesis. However, the precise roles of mitochondria-related genes (MRGs) and PCD-related genes (PCDRGs) in MM prognosis remain unclear. Methods Transcriptomic data from MM patients and healthy controls were analyzed to identify differentially expressed genes (DEGs). Candidate genes were selected by intersecting DEGs with curated lists of MRGs and PCDRGs. Univariate Cox, least absolute shrinkage and selection operator (LASSO), multivariate Cox, and stepwise regression analyses identified prognostic genes among the candidates. A risk model was constructed from these genes, and patients were stratified into high- and low-risk groups for survival analysis. Independent prognostic factors were incorporated into a nomogram to predict MM patient outcomes. Model performance was evaluated using calibration curves, receiver operating characteristic (ROC) analysis, and decision curve analysis (DCA). Finally, associations between prognostic genes and immune cell infiltration/drug responses were explored. Results 2,192 DEGs were detected between MM and control samples. 30 candidate genes were identified at the intersection of DEGs, 1,136 MRGs, and 1,548 PCDRGs. TRIAP1, TOMM7, PINK1, CHCHD10, PPIF, BCL2L1, and NDUFA13 were selected as prognostic genes. The risk model stratified patients into high- and low-risk groups with significantly different survival probabilities. Age, gender, ISS stage, and risk score were independent prognostic factors. The nomogram displayed good calibration and discriminative ability (AUC) in predicting survival, with clinical utility demonstrated by DCA. 9 immune cell types showed differential infiltration between MM and controls, with significant associations to risk scores and specific prognostic genes. 57 drugs, including nelarabine and vorinostat, were predicted to interact with the prognostic genes. Ultimately, qPCR in clinical samples from MM patients and healthy donors validated the expression levels of the seven key prognostic genes, corroborating the bioinformatic findings. Conclusion Seven genes (TRIAP1, TOMM7, PINK1, CHCHD10, PPIF, BCL2L1, NDUFA13) involved in mitochondrial function and PCD pathways were identified as prognostic markers in MM. These findings provide insights into MM biology and prognosis, highlighting potential therapeutic targets.
Collapse
Affiliation(s)
- Gongzhizi Gao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jiyu Miao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yachun Jia
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Xi’an Key Laboratory of Hematological Diseases, Xi’an, China
| |
Collapse
|
3
|
Garcia SA, Wilson K, Tang N, Tian H, Oichi T, Gunawardena AT, Chorny M, Alferiev IS, Herzenberg JE, Ng VY, Iwamoto M, Enomoto-Iwamoto M. Analysis of the Actions of RARγ Agonists on Growing Osteochondromas in a Mouse Model. Int J Mol Sci 2024; 25:7610. [PMID: 39062860 PMCID: PMC11277217 DOI: 10.3390/ijms25147610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
The actions of the retinoic acid nuclear receptor gamma (RARγ) agonist, palovarotene, on pre-existing osteochondromas were investigated using a mouse multiple osteochondroma model. This approach was based on the knowledge that patients often present to the clinic after realizing the existence of osteochondroma masses, and the findings from preclinical investigations are the effects of drugs on the initial formation of osteochondromas. Systemic administration of palovarotene, with increased doses (from 1.76 to 4.0 mg/kg) over time, fully inhibited tumor growth, keeping the tumor size (0.31 ± 0.049 mm3) similar to the initial size (0.27 ± 0.031 mm3, p = 0.66) while the control group tumor grew (1.03 ± 0.23 mm3, p = 0.023 to the drug-treated group). Nanoparticle (NP)-based local delivery of the RARγ agonist also inhibited the growth of osteochondromas at an early stage (Control: 0.52 ± 0.11 mm3; NP: 0.26 ± 0.10, p = 0.008). Transcriptome analysis revealed that the osteoarthritis pathway was activated in cultured chondrocytes treated with palovarotene (Z-score = 2.29), with the upregulation of matrix catabolic genes and the downregulation of matrix anabolic genes, consistent with the histology of palovarotene-treated osteochondromas. A reporter assay performed in cultured chondrocytes demonstrated that the Stat3 pathway, but not the Stat1/2 pathway, was stimulated by RARγ agonists. The activation of Stat3 by palovarotene was confirmed using immunoblotting and immunohistochemistry. These findings suggest that palovarotene treatment is effective against pre-existing osteochondromas and that the Stat3 pathway is involved in the antitumor actions of palovarotene.
Collapse
Affiliation(s)
- Sonia A. Garcia
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (S.A.G.); (K.W.); (N.T.); (H.T.); (T.O.); (V.Y.N.); (M.I.)
| | - Kimberly Wilson
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (S.A.G.); (K.W.); (N.T.); (H.T.); (T.O.); (V.Y.N.); (M.I.)
| | - Ningfeng Tang
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (S.A.G.); (K.W.); (N.T.); (H.T.); (T.O.); (V.Y.N.); (M.I.)
| | - Hongying Tian
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (S.A.G.); (K.W.); (N.T.); (H.T.); (T.O.); (V.Y.N.); (M.I.)
| | - Takeshi Oichi
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (S.A.G.); (K.W.); (N.T.); (H.T.); (T.O.); (V.Y.N.); (M.I.)
- Department of Orthopedics, Teikyo University School of Medicine, Tokyo 1738608, Japan
| | - Aruni T. Gunawardena
- Department of Biomechanics, Northeast College of Health Sciences, Seneca Falls, NY 13148, USA;
| | - Michael Chorny
- Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (M.C.); (I.S.A.)
| | - Ivan S. Alferiev
- Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (M.C.); (I.S.A.)
| | - John E. Herzenberg
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD 21215, USA;
| | - Vincent Y. Ng
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (S.A.G.); (K.W.); (N.T.); (H.T.); (T.O.); (V.Y.N.); (M.I.)
| | - Masahiro Iwamoto
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (S.A.G.); (K.W.); (N.T.); (H.T.); (T.O.); (V.Y.N.); (M.I.)
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (S.A.G.); (K.W.); (N.T.); (H.T.); (T.O.); (V.Y.N.); (M.I.)
| |
Collapse
|
4
|
Xu X, Feng J, Wang X, Zeng X, Luo Y, He X, Yang M, Lv T, Feng Z, Bao L, Zhao L, Huang D, Huang Y. Mitochondrial GRIM19 Loss Induces Liver Fibrosis through NLRP3/IL33 Activation via Reactive Oxygen Species/NF-кB Signaling. J Clin Transl Hepatol 2024; 12:539-550. [PMID: 38974954 PMCID: PMC11224902 DOI: 10.14218/jcth.2023.00562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/24/2024] [Accepted: 05/11/2024] [Indexed: 07/09/2024] Open
Abstract
Background and Aims Hepatic fibrosis (HF) is a critical step in the progression of hepatocellular carcinoma (HCC). Gene associated with retinoid-IFN-induced mortality 19 (GRIM19), an essential component of mitochondrial respiratory chain complex I, is frequently attenuated in various human cancers, including HCC. Here, we aimed to investigate the potential relationship and underlying mechanism between GRIM19 loss and HF pathogenesis. Methods GRIM19 expression was evaluated in normal liver tissues, hepatitis, hepatic cirrhosis, and HCC using human liver disease spectrum tissue microarrays. We studied hepatocyte-specific GRIM19 knockout mice and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein-9 (Cas9) lentivirus-mediated GRIM19 gene-editing in murine hepatocyte AML12 cells in vitro and in vivo. We performed flow cytometry, immunofluorescence, immunohistochemistry, western blotting, and pharmacological intervention to uncover the potential mechanisms underlying GRIM19 loss-induced HF. Results Mitochondrial GRIM19 was progressively downregulated in chronic liver disease tissues, including hepatitis, cirrhosis, and HCC tissues. Hepatocyte-specific GRIM19 heterozygous deletion induced spontaneous hepatitis and subsequent liver fibrogenesis in mice. In addition, GRIM19 loss caused chronic liver injury through reactive oxygen species (ROS)-mediated oxidative stress, resulting in aberrant NF-кB activation via an IKK/IкB partner in hepatocytes. Furthermore, GRIM19 loss activated NLRP3-mediated IL33 signaling via the ROS/NF-кB pathway in hepatocytes. Intraperitoneal administration of the NLRP3 inhibitor MCC950 dramatically alleviated GRIM19 loss-driven HF in vivo. Conclusions The mitochondrial GRIM19 loss facilitates liver fibrosis through NLRP3/IL33 activation via ROS/NF-кB signaling, providing potential therapeutic approaches for earlier HF prevention.
Collapse
Affiliation(s)
- Xiaohui Xu
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
- Department of Cardiology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Key Cardiovascular Specialty, Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing, China
| | - Jinmei Feng
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
- Department of Laboratory Medicine, Chongqing Western Hospital, Chongqing, China
| | - Xin Wang
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, China
| | - Xin Zeng
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Ying Luo
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Xinyu He
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Meihua Yang
- Departments of Neurology, Epilepsy Center, Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| | - Tiewei Lv
- Department of Cardiology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Key Cardiovascular Specialty, Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing, China
| | - Zijuan Feng
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Liming Bao
- Department of Clinical Pathology and Laboratory Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Li Zhao
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Daochao Huang
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Yi Huang
- Department of Cardiology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Key Cardiovascular Specialty, Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing, China
- Departments of Medicine (Oncology), Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
5
|
Yang L, Yang Y, Han X, Huang C, Wang Y, Jiang D, Chao L. GRIM19 deficiency aggravates metabolic disorder and ovarian dysfunction in PCOS. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167063. [PMID: 38360073 DOI: 10.1016/j.bbadis.2024.167063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders in women. Retinoid-interferon-induced mortality 19 (GRIM19) is a functional component of mitochondrial complex I that plays a role in cellular energy metabolism. However, the role of GRIM19 in the pathogenesis of PCOS is still unclear. OBJECTIVE To investigate the role of GRIM19 in the pathogenesis of PCOS. DESIGN We first measured the expression of GRIM19 in human granulosa cells (hGCs) from patients with and without PCOS (n = 16 per group), and then established a PCOS mouse model with WT and Grim19+/- mice for in vivo experiments. Glucose uptake-related genes RAC1 and GLUT4 and energy metabolism levels in KGN cells were examined in vitro by knocking down GRIM19 in the cell lines. Additionally, ovulation-related genes such as p-ERK1/2, HAS2, and PTX3 were also studied to determine their expression levels. RESULTS GRIM19 expression was reduced in hGCs of PCOS patients, which was negatively correlated with BMI and serum testosterone level. Grim19+/- mice with PCOS exhibited a markedly anovulatory phenotype and disturbed glycolipid metabolism. In vitro experiments, GRIM19 deficiency inhibited the RAC1/GLUT4 pathway, reducing insulin-stimulated glucose uptake in KGN cells. Moreover, GRIM19 deficiency induced mitochondrial dysfunction, defective glucose metabolism, and apoptosis. In addition, GRIM19 deficiency suppressed the expression of ovulation-related genes in KGN cells, which was regulated by dihydrotestosterone mediated androgen receptor. CONCLUSIONS GRIM19 deficiency may mediate ovulation and glucose metabolism disorders in PCOS patients. Our results suggest that GRIM19 may be a new target for diagnosis and treatment.
Collapse
Affiliation(s)
- Lin Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Yang Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Xiaojuan Han
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Chengzi Huang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Ying Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Danni Jiang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Lan Chao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
6
|
徐 小, 冯 金, 罗 颖, 何 昕, 臧 金, 黄 道. [Adeno-associated virus-mediated hepatocyte-specific NDUFA13 overexpression protects against CCl 4-induced liver fibrosis in mice by inhibiting hepatic NLRP3 activation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:201-209. [PMID: 38501404 PMCID: PMC10954519 DOI: 10.12122/j.issn.1673-4254.2024.02.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Indexed: 03/20/2024]
Abstract
OBJECTIVE To investigate the protective effect of NDUFA13 protein against acute liver injury and liver fibrosis in mice and explore the possible mechanisms. METHODS BALB/C mice (7 to 8 weeks old) were divided into normal group, CCl4 group, CCl4+AAV-NC group and CCl4+AAV-NDU13 group (n=18). Mouse models of liver fibrosis were established by intraperitoneal injection of CCl4 twice a week for 3, 5 or 7 weeks, and the recombinant virus AAV8-TBG-NC or AAV8-TBG-NDUFA13 was injected via the tail vein 7-10 days prior to CCl4 injection. After the treatments, pathological changes in the liver of the mice were observed using HE and Masson staining. Hepatic expression levels of NDUFA13 and α-SMA were detected with Western blotting, and the coexpression of NDUFA13 and NLRP3, TNF-α and IL-1β, and α-SMA and collagen Ⅲ was analyzed with immunofluorescence assay. RESULTS HE and Masson staining showed deranged liver architecture, necrotic hepatocytes and obvious inflammatory infiltration and collagen fiber deposition in mice with CCl4 injection (P < 0.001). NDUFA13 expression markedly decreased in CCl4-treated mice (P < 0.001), while a significant reduction in inflammatory aggregation and fibrosis was observed in mice with AAV-mediated NDUFA13 overexpression (P < 0.001). In CCl4+AAV-NDU13 group, immunofluorescence assay revealed markedly weakened activation of NLRP3 inflammasomes (P < 0.001), significantly decreased TNF-α and IL-1β secretion (P < 0.001), and inhibited hepatic stellate cell activation (P < 0.05) and collagen formation in the liver (P < 0.001). CONCLUSION Mitochondrial NDUFA13 overexpression in hepatocytes protects against CCl4- induced liver fibrosis in mice by inhibiting activation of NLRP3 signaling.
Collapse
Affiliation(s)
- 小惠 徐
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- 重庆医科大学附属儿童医院心内科//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室//国家临床心血管内科重点专科//重庆市卫生健康委儿童重要器官发育与疾病重点实验室,重庆 400014Department of Cardiology, Children's Hospital of Chongqing Medical University, Key Laboratory of Children's Important Organ Development and Diseases of Chongqing Municipal Health Commission, National Clinical Key Cardiovascular Specialty, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
| | - 金梅 冯
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- 重庆市九龙坡区第二人民医院检验科,重庆 400052Department of Laboratory Medicine, Second People's Hospital of Jiulongpo District, Chongqing 400052, China
| | - 颖 罗
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
| | - 昕觎 何
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
| | - 金宝 臧
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
| | - 道超 黄
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
| |
Collapse
|
7
|
Luo Y, Yang Y, Yang C, Li C, Hu R, Geng W, Kang X, Lin H. UBE3A and MCM6 synergistically regulate the proliferation and migration of lung adenocarcinoma cells. FEBS Open Bio 2023; 13:1756-1771. [PMID: 37454373 PMCID: PMC10476561 DOI: 10.1002/2211-5463.13675] [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: 02/07/2023] [Revised: 05/16/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023] Open
Abstract
Lung cancer is a leading cause of mortality worldwide and shows substantial clinical and biomolecular heterogeneity. Currently, specific therapeutic strategies are lacking, so effective drug targets are urgently needed. E6AP/UBE3A is a multifaceted ubiquitin ligase that controls various signaling pathways implicated in neurological diseases and various cancers; however, its role in lung cancer is incompletely understood. Here, MCM6 was identified as an interacting partner of E6AP using the yeast two-hybrid assay. MCM2 and MCM4 were then shown to interact with E6AP. E6AP knockout enhanced the ubiquitination of MCM2/4/6, suggesting that E6AP was not the E3 ubiquitin ligase for these three MCM proteins. Ablation of E6AP inhibited proliferation and migration, but had no significant effect on apoptosis in A549 and H1975 cells, and proliferation and migration inhibition was also observed in MCM6 knockdown cells. Furthermore, ablation of MCM6 and E6AP synergistically suppressed the proliferation and migration of A549 and H1975 cells. To verify the above findings in vivo, we established tumor models in nude mice and identified that the tumorigenicity of human lung adenocarcinoma (LUAD) cells was synergistically regulated by MCM6 and E6AP. Moreover, the expression levels of MCM6 and E6AP were higher in LUAD tissues than in adjacent tissues. Furthermore, the expression levels of MCM6 and E6AP were positively correlated in human LUAD samples. Thus, our study suggests that the interaction of E6AP and MCM proteins plays an important role in the progression of LUAD, which might offer potential therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Yanyan Luo
- Department of Pain, Wenzhou Key Laboratory of Perioperative MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityChina
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell Science, Chinese Academy of SciencesShanghaiChina
| | - Yun Yang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell Science, Chinese Academy of SciencesShanghaiChina
- School of MedicineGuizhou UniversityGuiyangChina
| | - Cong Yang
- Cancer Center, School of Medicine, Shanghai Tenth People's HospitalfTongji UniversityShanghaiChina
| | - Chuanyin Li
- Cancer Center, School of Medicine, Shanghai Tenth People's HospitalfTongji UniversityShanghaiChina
| | - Ronggui Hu
- Department of Pain, Wenzhou Key Laboratory of Perioperative MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityChina
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell Science, Chinese Academy of SciencesShanghaiChina
| | - Wujun Geng
- Department of Pain, Wenzhou Key Laboratory of Perioperative MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityChina
| | - Xianhui Kang
- Department of Pain, Wenzhou Key Laboratory of Perioperative MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityChina
- Department of Anesthesiology, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hai Lin
- Department of Pain, Wenzhou Key Laboratory of Perioperative MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityChina
| |
Collapse
|
8
|
Li Y, Liu X, Wan L, Han B, Ma S, Pan H, Wei J, Cui X. Metformin suppresses cardiac fibroblast proliferation under high-glucose conditions via regulating the mitochondrial complex I protein Grim-19 involved in the Sirt1/Stat3 signaling pathway. Free Radic Biol Med 2023; 206:1-12. [PMID: 37353174 DOI: 10.1016/j.freeradbiomed.2023.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/23/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023]
Abstract
Hyperglycemia associated with myocardial oxidative stress and fibrosis is the main cause of diabetic cardiomyopathy. Currently, no approved drug is available for preventing or treating diabetes-induced cardiac fibrosis. Metformin has been reported to improve glycemic control and ameliorate diabetic cardiomyopathy. This study aimed to investigate the effects and mechanism of metformin on diabetes-induced cardiac fibrosis and high glucose-induced proliferation of cardiac fibroblasts (CFs). In this study, db/db mice were treated with metformin [250 mg/kg⋅d, gavage]. CFs were cultured in high-glucose medium to mimic an in vitro diabetes model and then subjected to treatment with or without metformin. Cardiac fibrosis was analyzed using immunohistochemistry, Masson's trichrome staining, and Western blot analysis. Cell Counting Kit-8 (CCK-8) assays and cell colony formation assays were used to examine cell proliferation capacity. Transwell and scratch-wound assays were used to detect the migration ability of CFs. Retinoid-interferon-induced mortality-19 (Grim-19), sirtuin1 (Sirt1), and signal transducer and activator of transcription 3 (Stat3) were detected using Western blot analysis. The genes downstream of the Stat3 pathway were detected using quantitative reverse transcription PCR (qRT‒PCR). Metformin treatment markedly attenuated cardiac fibrosis in db/db mice and the proliferation and migration of CFs under high-glucose conditions. Mechanistically, we found an intersection between metformin and Grim-19 using bioinformatics. Metformin was found to suppress the expression of p-Stat3 and elevate the expression of mitochondrial complex I protein Grim-19 and Sirt1, thus inhibiting the proliferation and migration of CFs under high-glucose conditions. Our data suggested that metformin inhibited the proliferation and migration of CFs by regulating the expression of mitochondrial complex I Grim-19 protein involved in the Sirt1/Stat3 signaling pathway under high-glucose conditions, thus providing new ideas for treating diabetes-induced cardiac fibrosis.
Collapse
Affiliation(s)
- Yongguang Li
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Xiangdong Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200000, People's Republic of China
| | - Lili Wan
- Division of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Beibei Han
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Shixin Ma
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Hongyuan Pan
- Saint Paul's School, 325 Pleasant Street, Concord, NH, 03301, USA
| | - Junbo Wei
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 Yishan Road, Shanghai, 200233, People's Republic of China; Department of Cardiology, Renhe Hospital, 1999 Changjiang West Road, Shanghai, 200431, People's Republic of China.
| | - Xiaofang Cui
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
9
|
Wagner ML, Ammann A, Piraino G, Wolfe V, O’Connor M, Lahni P, Ziady A, Zingarelli B. PROTECTIVE EFFECTS OF HUMANIN-G IN HEMORRHAGIC SHOCK IN FEMALE MICE VIA AMPKα1-INDEPENDENT MECHANISMS. Shock 2023; 60:64-74. [PMID: 37079467 PMCID: PMC10523894 DOI: 10.1097/shk.0000000000002134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
ABSTRACT Introduction: Despite therapeutic advances in hemorrhagic shock, mortality from multiple organ failure remains high. We previously showed that the α1 subunit of AMP-activated protein kinase (AMPK), a crucial regulator of mitochondrial function, exerts a protective role in hemorrhagic shock. Humanin is a mitochondrial peptide with cytoprotective properties against cellular stress. Here, we investigated whether AMPKα1 influences systemic levels of endogenous humanin in hemorrhagic shock and whether treatment with the synthetic analog humanin-G affords beneficial effects. Methods: AMPKα1 wild-type (WT) and knockout (KO) female mice were subjected to hemorrhagic shock followed by resuscitation with blood and lactated Ringer's solution. In short-term studies, mice were treated with humanin-G or vehicle and sacrificed at 3 h after resuscitation; in survival studies, mice were treated with PEGylated humanin-G and monitored for 7 days. Results: Compared with the vehicle WT group, KO mice exhibited severe hypotension, cardiac mitochondrial damage, and higher plasma levels of Th17 cytokines but had similar lung injury and similar plasma elevation of endogenous humanin. Treatment with humanin-G improved lung injury, mean arterial blood pressure, and survival in both WT and KO mice, without affecting systemic cytokine or humanin levels. Humanin-G also ameliorated cardiac mitochondrial damage and increased adenosine triphosphate levels in KO mice. Beneficial effects of humanin-G were associated with lung cytoplasmic and nuclear activation of the signal transducer and activator of transcription-3 (STAT3) in AMPKα1-independent manner with marginal or no effects on mitochondrial STAT3 and complex I subunit GRIM-19. Conclusions: Our data indicate that circulating levels of humanin increase during hemorrhagic shock in AMPKα1-independent fashion as a defense mechanism to counteract metabolic derangement and that administration of humanin-G affords beneficial effects through STAT3 activation even in the absence of a functional AMPKα1.
Collapse
Affiliation(s)
- Monica L. Wagner
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati Ohio, USA
| | - Allison Ammann
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati Ohio, USA
| | - Giovanna Piraino
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati Ohio, USA
| | - Vivian Wolfe
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati Ohio, USA
| | - Michael O’Connor
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati Ohio, USA
| | - Patrick Lahni
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati Ohio, USA
| | - Assem Ziady
- Division of Bone Marrow Transplantation & Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Ohio, USA
| | - Basilia Zingarelli
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Ohio, USA
| |
Collapse
|
10
|
Smith S, Lopez S, Kim A, Kasteri J, Olumuyide E, Punu K, de la Parra C, Sauane M. Interleukin 24: Signal Transduction Pathways. Cancers (Basel) 2023; 15:3365. [PMID: 37444474 PMCID: PMC10340555 DOI: 10.3390/cancers15133365] [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: 05/25/2023] [Revised: 06/16/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Interleukin 24 is a member of the IL-10 family with crucial roles in antitumor, wound healing responses, host defense, immune regulation, and inflammation. Interleukin 24 is produced by both immune and nonimmune cells. Its canonical pathway relies on recognition and interaction with specific Interleukin 20 receptors in the plasma membrane and subsequent cytoplasmic Janus protein tyrosine kinases (JAK)/signal transducer and activator of the transcription (STAT) activation. The identification of noncanonical JAK/STAT-independent signaling pathways downstream of IL-24 relies on the interaction of IL-24 with protein kinase R in the cytosol, respiratory chain proteins in the inner mitochondrial membrane, and chaperones such as Sigma 1 Receptor in the endoplasmic reticulum. Numerous studies have shown that enhancing or inhibiting the expression of Interleukin 24 has a therapeutic effect in animal models and clinical trials in different pathologies. Successful drug targeting will require a deeper understanding of the downstream signaling pathways. In this review, we discuss the signaling pathway triggered by IL-24.
Collapse
Affiliation(s)
- Simira Smith
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
| | - Sual Lopez
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
| | - Anastassiya Kim
- Ph.D. Program in Biology, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; (A.K.); (C.d.l.P.)
| | - Justina Kasteri
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
| | - Ezekiel Olumuyide
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
| | - Kristian Punu
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
| | - Columba de la Parra
- Ph.D. Program in Biology, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; (A.K.); (C.d.l.P.)
- Department of Chemistry, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA
| | - Moira Sauane
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
- Ph.D. Program in Biology, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; (A.K.); (C.d.l.P.)
| |
Collapse
|
11
|
Li L, Li Y, Yang J, Xie X, Chen H. The immune responses to different Uropathogens call individual interventions for bladder infection. Front Immunol 2022; 13:953354. [PMID: 36081496 PMCID: PMC9445553 DOI: 10.3389/fimmu.2022.953354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Urinary tract infection (UTI) caused by uropathogens is the most common infectious disease and significantly affects all aspects of the quality of life of the patients. However, uropathogens are increasingly becoming antibiotic-resistant, which threatens the only effective treatment option available-antibiotic, resulting in higher medical costs, prolonged hospital stays, and increased mortality. Currently, people are turning their attention to the immune responses, hoping to find effective immunotherapeutic interventions which can be alternatives to the overuse of antibiotic drugs. Bladder infections are caused by the main nine uropathogens and the bladder executes different immune responses depending on the type of uropathogens. It is essential to understand the immune responses to diverse uropathogens in bladder infection for guiding the design and development of immunotherapeutic interventions. This review firstly sorts out and comparatively analyzes the immune responses to the main nine uropathogens in bladder infection, and summarizes their similarities and differences. Based on these immune responses, we innovatively propose that different microbial bladder infections should adopt corresponding immunomodulatory interventions, and the same immunomodulatory intervention can also be applied to diverse microbial infections if they share the same effective therapeutic targets.
Collapse
Affiliation(s)
- Linlong Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Yangyang Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jiali Yang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- *Correspondence: Xiang Xie, ; Huan Chen,
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- *Correspondence: Xiang Xie, ; Huan Chen,
| |
Collapse
|
12
|
Huang Y, Zhao Y, Liu H, Yang Y, Cheng L, Deng X, Chao L. Decreased expression of GRIM-19 induces autophagy through the AMPK/ULK1 signaling pathway during adenomyosis. Biol Reprod 2022; 107:956-966. [PMID: 35908189 DOI: 10.1093/biolre/ioac151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 06/07/2022] [Indexed: 11/14/2022] Open
Abstract
The processes underlying adenomyosis are similar to those of tumor metastasis, and it is defined as progressive invasion by the endometrium and the subsequent creation of ectopic lesions. GRIM-19 regulates cell death via the mitochondrial respiratory chain. Stress following oxygen deprivation can induce tumor cell autophagy, leading to cell invasion and migration. Here, we revealed that GRIM-19 negatively regulates autophagy, and, at least in adenomyosis, decreased expression of GRIM-19 is accompanied by an increased level of autophagy and 5'-adenosine monophosphate-activated protein kinase-Unc-51 like autophagy activating kinase 1 (AMPK-ULK1) activation. Upregulation of GRIM-19 expression in human primary endometrial cells and ISHIKAWA cells inhibits autophagy via the AMPK-ULK1 pathway and helps control cell invasion and migration. In addition, we also identified increased expression of AMPK and ULK1, and higher levels of autophagy in the uterine tissues of GRIM-19+/- mice. Importantly, the function of the GRIM-19-AMPK-ULK1 axis in regulating autophagy in adenomyosis is similar to that of tumor tissues, which may help elucidate the regulation of adenomyosis tumor-like behavior, and is expected to help identify novel targets for the diagnosis and treatment of adenomyosis.
Collapse
Affiliation(s)
- YuFei Huang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Yue Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - HaoRan Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Yang Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - LaiYang Cheng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - XiaoHui Deng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Lan Chao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| |
Collapse
|
13
|
Lin C, Wang J, Wang Y, Chen C, Gao X. The postoperative cognitive dysfunction induced by central inflammation with possible involvement of the gut-brain axis. Clinics (Sao Paulo) 2022; 77:100104. [PMID: 36137346 PMCID: PMC9493054 DOI: 10.1016/j.clinsp.2022.100104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 06/06/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Postoperative cognitive dysfunction is widely recognized as severe postoperative central nervous dysfunction and has a significant impact on the 'patient's physical and mental health. METHODS Postoperative models of tibial fracture in aged rats were established, including the control group, model group, CCL11 protein injection group, and saline injection group. Morris water maze test was used to detect the behavioral characteristics of rats. Enzyme-Linked Immunosorbent Assay was used or determine the content of CCL11 and CXCL10. Immunofluorescence staining was used to detect the distribution of CD14+CD163+macrophages in colon tissues and CD11b+CCR3+microglia cells in hippocampal tissues. Western blot analyzed NOX1 and STAT3 expression in hippocampus tissues. RESULTS Water maze test results confirmed severe cognitive impairment in CCL11 rats. The content of CCL11 and CXCL10 in the CCL11 group was much higher than that of the model group. The distribution of macrophage and microglia cells in the CCL11 model group was greater than that in the model group and the saline group. The expression of NOX1 and STAT3 in the CCL11 group was higher compared with the model group. CONCLUSION Abnormal macrophage function and excessive CCL11 secretion were observed in the rats with lower limb fractures after surgery. Postoperative central inflammation in rats with lower limb fracture induced postoperative cognitive dysfunction through the gut-brain axis molecular mechanism.
Collapse
Affiliation(s)
- Chuantao Lin
- Department of Anesthesiology, Fujian Maternity and Child Health Hospital; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian, China
| | - Jing Wang
- Department of Anesthesiology, Fujian Maternity and Child Health Hospital; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian, China
| | - Yuping Wang
- Department of Anesthesiology, Fujian Maternity and Child Health Hospital; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian, China
| | - Chanjuan Chen
- Department of Anesthesiology, Fujian Maternity and Child Health Hospital; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian, China
| | - Xiang Gao
- Department of Anesthesiology, Fujian Maternity and Child Health Hospital; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian, China.
| |
Collapse
|
14
|
Yang Y, Liu H, Zhao Y, Geng C, Chao L, Hao A. Grim-19 deficiency promotes decidual macrophage autophagy in recurrent spontaneous abortion. Front Endocrinol (Lausanne) 2022; 13:1023194. [PMID: 36387896 PMCID: PMC9641028 DOI: 10.3389/fendo.2022.1023194] [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: 08/19/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Dysregulation of decidual macrophages leads to the occurrence of recurrent spontaneous abortion (RSA). However, the role of macrophages in RSA occurrence remains unclear. In this study, we found that the expression of Grim-19 was decreased, and the expression of autophagy related proteins Beclin1, LC3B II/I and BNIP3 was markedly upregulated in decidual macrophages of RSA patients compared with the normal pregnancy group. Furthermore, we demonstrated that downregulation of GRIM-19 increased the expression of autophagy related proteins Beclin1, LC3B II/I, BNIP3 and the proinflammatory cytokines IL1B, IL6 and TNFa in uterine mononuclear cells of GRIM-19+/- mice. The proportion of CD45+CD11b+F4/80+LC3B+ cells in GRIM-19+/- mouse uteri was significantly higher than that in WT mouse uteri. In addition, we confirmed that inhibition of Grim-19 by siRNA enhanced the expression of autophagy related proteins in RAW264.7 cells and THP-1 cells. More importantly, downregulation of Grim-19 in RAW264.7 cells promoted the release of proinflammatory cytokines and promoted phagocytic activity, which could be reversed by autophagy blockade. For THP-1-derived macrophages, the results of RNA-seq suggested that Grim-19 mainly modulates immune and inflammatory-related pathways, leading to cytokine production, and thus contributing to inflammation. Therefore, our data reveal that Grim-19 deficiency influences macrophage function, characterized by enhanced proinflammatory cytokines and phagocytic activity, and this might be regulated by autophagy. This may represent a novel mechanism for the occurrence of RSA.
Collapse
Affiliation(s)
- Yang Yang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Haoran Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Yue Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Chen Geng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Lan Chao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Aijun Hao
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Aijun Hao,
| |
Collapse
|
15
|
Kurane T, Matsunaga T, Ida T, Sawada K, Nishimura A, Fukui M, Umemura M, Nakayama M, Ohara N, Matsumoto S, Akaike T, Matsuzaki G, Takaesu G. GRIM-19 is a target of mycobacterial Zn 2+ metalloprotease 1 and indispensable for NLRP3 inflammasome activation. FASEB J 2021; 36:e22096. [PMID: 34907600 DOI: 10.1096/fj.202101074rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 11/11/2022]
Abstract
Tuberculosis is a communicable disease caused by Mycobacterium tuberculosis which primarily infects macrophages and establishes intracellular parasitism. A mycobacterial virulence factor Zn2+ metalloprotease 1 (Zmp1) is known to suppress interleukin (IL)-1β production by inhibiting caspase-1 resulting in phagosome maturation arrest. However, the molecular mechanism of caspase-1 inhibition by Zmp1 is still elusive. Here, we identified GRIM-19 (also known as NDUFA13), an essential subunit of mitochondrial respiratory chain complex I, as a novel Zmp1-binding protein. Using the CRISPR/Cas9 system, we generated GRIM-19 knockout murine macrophage cell line J774.1 and found that GRIM-19 is essential for IL-1β production during mycobacterial infection as well as in response to NLRP3 inflammasome-activating stimuli such as extracellular ATP or nigericin. We also found that GRIM-19 is required for the generation of mitochondrial reactive oxygen species and NLRP3-dependent activation of caspase-1. Loss of GRIM-19 or forced expression of Zmp1 resulted in a decrease in mitochondrial membrane potential. Our study revealed a previously unrecognized role of GRIM-19 as an essential regulator of NLRP3 inflammasome and a molecular mechanism underlying Zmp1-mediated suppression of IL-1β production during mycobacterial infection.
Collapse
Affiliation(s)
- Tomomi Kurane
- Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Tetsuro Matsunaga
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoaki Ida
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuko Sawada
- Molecular Microbiology Group, Tropical Biosphere Research Center, University of the Ryukyus, Okinawa, Japan
| | - Akira Nishimura
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masayuki Fukui
- Molecular Microbiology Group, Tropical Biosphere Research Center, University of the Ryukyus, Okinawa, Japan
| | - Masayuki Umemura
- Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan.,Molecular Microbiology Group, Tropical Biosphere Research Center, University of the Ryukyus, Okinawa, Japan.,Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Masaaki Nakayama
- Department of Oral Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Naoya Ohara
- Department of Oral Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Sohkichi Matsumoto
- Department of Bacteriology, Niigata University School of Medicine, Niigata, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Goro Matsuzaki
- Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan.,Molecular Microbiology Group, Tropical Biosphere Research Center, University of the Ryukyus, Okinawa, Japan.,Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Giichi Takaesu
- Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan.,Molecular Microbiology Group, Tropical Biosphere Research Center, University of the Ryukyus, Okinawa, Japan.,Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| |
Collapse
|
16
|
Wang D, Wei X, Chen X, Wang Q, Zhang J, Kalvakolanu DV, Guo B, Zhang L. GRIM-19 inhibits proliferation and induces apoptosis in a p53-dependent manner in colorectal cancer cells through the SIRT7/PCAF/MDM2 axis. Exp Cell Res 2021; 407:112799. [PMID: 34461110 DOI: 10.1016/j.yexcr.2021.112799] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/16/2021] [Accepted: 08/25/2021] [Indexed: 10/20/2022]
Abstract
Colorectal cancer (CRC) is the leading deadly cancer worldwide. Gene associated with retinoid-IFN-induced mortality-19 (GRIM-19), a novel tumor suppressor, has been reported to be expressed at low levels in human CRC. However, the role of GRIM-19 in CRC progression and the corresponding detailed mechanisms are unclear. The results of this study indicated that GRIM-19 expression is related to CRC progression. Overexpression of GRIM-19 was found to inhibit CRC cell proliferation and induce apoptosis in vitro and in vivo. Our results demonstrated that GRIM-19 suppresses CRC through posttranslational regulation of p53, in which SIRT7 is activated by GRIM-19 and triggers PCAF-mediated MDM2 ubiquitination, eventually stabilizing the p53 protein. We also observed that GRIM-19 enhances the effect of oxaliplatin against CRC. In conclusion, GRIM-19 plays an important role in CRC development and is a potential biomarker and therapeutic target for clinical treatment of CRC.
Collapse
Affiliation(s)
- Ding Wang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, PR China
| | - Xiaodong Wei
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, PR China
| | - Xuyang Chen
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, PR China
| | - Qian Wang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, PR China
| | - Jinghua Zhang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, PR China
| | - Dhan V Kalvakolanu
- Greenebaum NCI Comprehensive Cancer Center, Department of Microbiology and Immunology University of Maryland School Medicine, Baltimore, MD, USA
| | - Baofeng Guo
- Department of Plastic Surgery, China-Japan Union Hospital of Jilin University, Changchun, PR China.
| | - Ling Zhang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, PR China.
| |
Collapse
|
17
|
Xu X, Zeng X, Li R, Feng J, Huang D, Huang Y. [Mechanism of hepatocyte mitochondrial NDUFA13 deficiency-induced liver fibrogenesis: the role of abnormal hepatic stellate cell activation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:529-535. [PMID: 33963711 DOI: 10.12122/j.issn.1673-4254.2021.04.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the role of hepatocyte mitochondrial NDUFA13 loss in the liver fibrogenesis in mice and explore the possible mechanisms. OBJECTIVE We used liver-specific NDUFA13 heterozygous knockout mouse models (NDUFA13fl/-; Alb-Cre) established previously by intercrossing NDUFA13fl/fl and Alb-Cre mice, with their littermate control NDUFA13fl/fl mice as the control (n=8). The mice were euthanized at the age of 4 weeks and 2 years, and the liver tissues were collected for HE and Masson staining to observe the pathological changes and fibrosis phenotypes. Western blotting was performed to detect the expression of NDUFA13 protein in the liver tissues, and the infiltration of F4/80+ macrophages and the expressions of TGF-β1, TNF-α and IL-1β were analyzed by immunofluorescence assay. The expression levels of α-SMA, matrix metalloproteinase-9 (MMP-9) and tissue inhibitor of matrix metalloproteases 1 (TIMP-1), collagen-Ⅰ and collagen-Ⅲ were assayed by immunohistochemistry. OBJECTIVE HE and Masson staining showed obvious inflammatory infiltration but no significant fibrosis in the liver tissues of 4-week-old NDUFA13fl/- mice, but severe liver damage with massive fibrosis was observed in 2-year-old NDUFA13fl/- mice. NDUFA13 expression in 2-year-old NDUFA13fl/- mice markedly decreased compared with that in the control NDUFA13fl/fl mice as shown by Western blotting (P < 0.05). Immunohistochemistry showed obvious infiltration of F4/80+ macrophages in the liver tissue with a large amount of TGF-β1 production (P < 0.05) and TNF-α and IL-1β secretions in NDUFA13fl/- mice (P < 0.05). NDUFA13 knockout obviously promoted α-SMA expression (P < 0.05) and collagen-Ⅰ and collagen-Ⅲ deposition (P < 0.05) while significantly decreased MMP-9 and increased TIMP-1 expression in the liver (P < 0.05). OBJECTIVE Hepatocytes-specific NDUFA13 deficiency can trigger spontaneous and chronic liver fibrosis phenotypes in mice probably in association with abnormal activation of hepatic stellate cells induced by macrophages and inflammatory factors.
Collapse
Affiliation(s)
- X Xu
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| | - X Zeng
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| | - R Li
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| | - J Feng
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| | - D Huang
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| | - Y Huang
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| |
Collapse
|
18
|
Knockdown of Gastrin Promotes Apoptosis of Gastric Cancer Cells by Decreasing ROS Generation. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5590037. [PMID: 33937399 PMCID: PMC8062189 DOI: 10.1155/2021/5590037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/01/2021] [Indexed: 12/27/2022]
Abstract
Overexpressed gastrin is reported to promote oncogenesis and development of gastric cancer by inhibiting apoptosis of cancer cells; however, the underlying mechanism remains unclear. Our study is aimed at revealing the mechanism underlying the effect of gastrin on apoptosis of gastric cancer cells. Gastrin-interfering cell line was constructed by stably transfecting gastrin-specific pshRNA plasmid to gastric cancer cell line BGC-823. Then, differentially expressed proteins between untreated BGC-823 and gastrin-interfering BGC-823 cell lines were detected by the iTRAQ technique. GO and KEGG analysis was used to analyze the differentially expressed genes that code these differentially expressed proteins. The Annexin V-FITC staining assay was used to detect gastric cancer cell apoptosis. The DCFH-DA fluorescent probe staining assay was used to measure intracellular ROS. Mitochondrial membrane potential was detected by flow cytometry. Western blot was used to analyze the mitochondria respiratory chain proteins and apoptosis-related proteins. A total of 107 differentially expressed proteins were identified by iTRAQ. GO and KEGG analysis showed that proteins coded by the corresponding differentially expressed genes were mainly enriched in the mitochondrial oxidative respiratory chain, and the expression of three proteins (COX17, COX5B, ATP5J) was upregulated. The three proteins with higher scores were verified by Western blot. The apoptosis rate of the gastrin knockdown cancer cell was significantly increased; meanwhile, gastrin knockdown leads to increase of membrane potential and decrease of intracellular ROS production. Additionally, Bax was significantly increased, whereas NF-κB-p65 and Bcl-2 were downregulated after knockdown of gastrin. Concomitantly, pretreatment with NAC reversed the effect of gastrin on the Bax and Bcl-2 expression. Gastrin promotes the production of ROS from mitochondria, activates NF-κB, and inhibits apoptosis via modulating the expression level of Bcl-2 and Bax.
Collapse
|
19
|
Xu X, Li R, Zeng X, Wang X, Xue B, Huang D, Huang Y. [Pathogenic role of NDUFA13 inactivation in spontaneous hepatitis in mice and the mechanism]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:55-63. [PMID: 33509753 DOI: 10.12122/j.issn.1673-4254.2021.01.07] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate the role of NDUFA13 inactivation in the pathogenesis of spontaneous hepatitis in mice and explore the possible mechanisms. METHODS Hepatocyte-specific NDUFA13 knockout (NDUFA13fl/-) mice were generated by intercrossing NDUFA13fl/fl and Alb-Cre mice based on Cre/loxP transgenic technology, and tail and liver DNA of the mice was genotyped by PCR analysis. Ten NDUFA13fl/- mice and 10 littermate control NDUFA13fl/fl mice were housed, and in each group, 5 mice were euthanized at the age of 4 weeks and the other 5 at two years for pathological examination of the liver tissues with HE staining. Immunohistochemistry was used to verify the expression levels of NDUFA13, NF-κB/p65, NF-κB/p-p65 and inflammasome NLRP3. The total intracellular ROS and mitochondrial ROS levels were measured with a ROS staining kit. The expressions of the inflammatory cell markers (CD45, MPO, and F4/80) and inflammatory cytokines (IL1β and IL33) in the liver were detected with immunohistochemistry and immunofluorescence assay. RESULTS Liver-specific NDUFA13 heterozygous knockout mice were successfully constructed as verified by PCR results. HE staining revealed severe liver damage in both 4- week-old and 2-year-old NDUFA13fl/- mice as compared with their littermate controls. Immunohistochemistry showed a significant decrease of NDUFA13 expression in both 4-week-old and 2-year-old NDUFA13fl/- mice (P < 0.05). The expression levels of NF-κB signals p65, p-p65 and NLRP3 inflammasomes were all significantly increased in NDUFA13fl/- mice (P < 0.05). The total intracellular ROS and mitochondrial ROS levels in NDUFA13fl/- mice were also significantly increased. NDUFA13 knockout obviously promoted the expression of the inflammatory cell markers (CD45, MPO and F4/80) and the secretion of IL-1β and IL-33 in the liver tissue of the mice (P < 0.05). CONCLUSIONS Hepatocytes-specific NDUFA13 ablation can trigger spontaneous hepatitis in mice possibly mediated by the activation of ROS/NF-κB/NLRP3 signaling.
Collapse
Affiliation(s)
- Xiaohui Xu
- Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Infection and Immunity// Key Laboratory of Child Development and Disorders of Ministry of Education//National Clinical Research Center for Child Health and Disorders//China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Rui Li
- Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Infection and Immunity// Key Laboratory of Child Development and Disorders of Ministry of Education//National Clinical Research Center for Child Health and Disorders//China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Xin Zeng
- Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Infection and Immunity// Key Laboratory of Child Development and Disorders of Ministry of Education//National Clinical Research Center for Child Health and Disorders//China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Xin Wang
- Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Infection and Immunity// Key Laboratory of Child Development and Disorders of Ministry of Education//National Clinical Research Center for Child Health and Disorders//China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Bingqian Xue
- Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Infection and Immunity// Key Laboratory of Child Development and Disorders of Ministry of Education//National Clinical Research Center for Child Health and Disorders//China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Daochao Huang
- Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Infection and Immunity// Key Laboratory of Child Development and Disorders of Ministry of Education//National Clinical Research Center for Child Health and Disorders//China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Yi Huang
- Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Infection and Immunity// Key Laboratory of Child Development and Disorders of Ministry of Education//National Clinical Research Center for Child Health and Disorders//China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| |
Collapse
|
20
|
Moon J, Lee SH, Lee SY, Ryu J, Jhun J, Choi J, Kim GN, Roh S, Park SH, Cho ML. GRIM-19 Ameliorates Multiple Sclerosis in a Mouse Model of Experimental Autoimmune Encephalomyelitis with Reciprocal Regulation of IFNγ/Th1 and IL-17A/Th17 Cells. Immune Netw 2020; 20:e40. [PMID: 33163248 PMCID: PMC7609166 DOI: 10.4110/in.2020.20.e40] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/05/2020] [Accepted: 09/23/2020] [Indexed: 01/14/2023] Open
Abstract
The protein encoded by the Gene Associated with Retinoid-Interferon-Induced Mortality-19 (GRIM-19) is located in the mitochondrial inner membrane and is homologous to the NADH dehydrogenase 1-alpha subcomplex subunit 13 of the electron transport chain. Multiple sclerosis (MS) is a demyelinating disease that damages the brain and spinal cord. Although both the cause and mechanism of MS progression remain unclear, it is accepted that an immune disorder is involved. We explored whether GRIM-19 ameliorated MS by increasing the levels of inflammatory cytokines and immune cells; we used a mouse model of experimental autoimmune encephalomyelitis (EAE) to this end. Six-to-eight-week-old male C57BL/6, IFNγ-knockout (KO), and GRIM-19 transgenic mice were used; EAE was induced in all strains. A GRIM-19 overexpression vector (GRIM19 OVN) was electrophoretically injected intravenously. The levels of Th1 and Th17 cells were measured via flow cytometry, immunofluorescence, and immunohistochemical analysis. IL-17A and IFNγ expression levels were assessed via ELISA and quantitative PCR. IL-17A expression decreased and IFNγ expression increased in EAE mice that received injections of the GRIM19 OVN. GRIM-19 transgenic mice expressed more IFNγ than did wild-type mice; this inhibited EAE development. However, the effect of GRIM-19 overexpression on the EAE of IFNγ-KO mice did not differ from that of the empty vector. GRIM-19 expression was therapeutic for EAE mice, elevating the IFNγ level. GRIM-19 regulated the Th17/Treg cell balance.
Collapse
Affiliation(s)
- Jeonghyeon Moon
- Laboratory of Immune Network, Conversant Research Consortium in Immunologic Disease, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, BK21 PLUS Dental Life Science, Seoul National University School of Dentistry, Seoul 08826, Korea
| | - Seung Hoon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Seon-Yeong Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jaeyoon Ryu
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jooyeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - JeongWon Choi
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Gyoung Nyun Kim
- College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, BK21 PLUS Dental Life Science, Seoul National University School of Dentistry, Seoul 08826, Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Mi-La Cho
- Laboratory of Immune Network, Conversant Research Consortium in Immunologic Disease, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Department of Medical Lifescience, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
21
|
Wang B, Yang Y, Deng X, Ban Y, Chao L. Interaction of M2 macrophages and endometrial cells induces downregulation of GRIM-19 in endometria of adenomyosis. Reprod Biomed Online 2020; 41:790-800. [PMID: 32896475 DOI: 10.1016/j.rbmo.2020.04.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 04/11/2020] [Accepted: 04/28/2020] [Indexed: 11/30/2022]
Abstract
RESEARCH QUESTION Does the aggregation of M2 macrophages affect the expression of gene associated with retinoid-interferon-induced mortality 19 (GRIM-19) in adenomyosis? DESIGN Endometrial tissues were collected from patients with (n = 15) and without (n = 15) adenomyosis. Tissues were analysed for GRIM-19 and toll-like receptor 4 (TLR4) expression by immunohistochemistry and western blotting. Apoptosis was analysed by TdT (terminal deoxynucleotidyl transferase)-mediated dUDP nick-end labelling (TUNEL) assay. Human endometrial stromal cells (HESC) were transfected with GRIM-19 small interfering RNA (SiRNA) to knockdown GRIM-19 expression. The HESC were co-cultured with M2 macrophages to detect the influence of M2 macrophages in HESC cells. Analyses included GRIM-19, caspase-3 and TLR4 expression by western blotting, and GRIM-19 and TLR4 by quantitative real-time polymerase chain reaction. Apoptosis was measured by flow cytometry and TUNEL assay. Cell proliferation (Cell Counting Kit-8 assay) and migration assays were carried out. RESULTS The expression of GRIM-19 was significantly lower in adenomyosis lesions compared with controls (P < 0.001). Deficiency of GRIM-19 induced by siRNA decreased apoptosis and increased proliferation and migration in HESC. A significant decrease in GRIM-19 expression occurred in HESC after co-culture with M2 macrophages (P = 0.018). After co-culture with M2 macrophage, apoptosis decreased and proliferation and cell invasion in HESC increased. Protein (P = 0.006) and mRNA (P = 0.013) expression of TLR4 in HESC also reduced after this co-culture. Up-regulation of GRIM-19 occurred in HESC treated with the activator TLR4 (P = 0.016). Up-regulation of GRIM-19 was significantly reversed in cells treated with the TLR4 inhibitor (P = 0.011). CONCLUSION M2 macrophages may be involved in regulating the expression of GRIM-19 partly through the TLR4 signalling axis in adenomyosis.
Collapse
Affiliation(s)
- Bingyu Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Yang Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Xiaohui Deng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Yanli Ban
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Lan Chao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
22
|
Sadhukhan P, Sil PC. The regulation of intracellular redox homeostasis in cancer progression and its therapy. Pathology 2020. [DOI: 10.1016/b978-0-12-815972-9.00010-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
23
|
Xu Y, Wang X, Guo B, Wang D, Kalvakolanu DV, Chen X, Tang J, Zhang L, Yang Q. Nonviral Delivery of GRIM-19 Gene Inhibits Tumor Growth with Reduced Local and Systemic Complications. Hum Gene Ther 2019; 30:1419-1430. [DOI: 10.1089/hum.2019.134] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Yang Xu
- Departments of Pathogenobiology and Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xinghuo Wang
- Department of Polymer Science, College of Chemistry, Jilin University, Changchun, China
| | - Baofeng Guo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Ding Wang
- Departments of Pathogenobiology and Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Dhan V. Kalvakolanu
- Greenebaum Cancer Center, Department of Microbiology and Immunology, Molecular Biology Program, University of Maryland School Medicine, Baltimore, Maryland
| | - Xuyang Chen
- Departments of Pathogenobiology and Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jun Tang
- Departments of Pathogenobiology and Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
- Department of Polymer Science, College of Chemistry, Jilin University, Changchun, China
| | - Ling Zhang
- Departments of Pathogenobiology and Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Qing Yang
- Departments of Pathogenobiology and Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
24
|
Lin H, Lin T, Lin J, Yang M, Shen Z, Liu H, Zou Z, Zheng Z. Inhibition of miR-423-5p suppressed prostate cancer through targeting GRIM-19. Gene 2019; 688:93-97. [PMID: 30415005 DOI: 10.1016/j.gene.2018.11.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/07/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To determine the effect of miR-423-5p on the progression of prostate cancer (PC). METHODS miR-423-5p and GRIM-19 expressions were detected by qRT-PCR and western blot. PC cell proliferation was measured by MTT assay. PC cell apoptosis was detected by flow cytometry. Dual luciferase reporter assay was used to confirm the interaction between miR-423-5p and GRIM-19. RESULTS Compared with normal prostate tissues and prostate epithelial cell HPrEC, miR-423-5p was up-regulated in human PC tissues and PC3 cells, whereas GRIM-19 expression was decreased. Inhibition of miR-423-5p suppressed PC3 cell proliferation, promoted PC3 cell apoptosis, and decreased anti-apoptosis protein BCL-2 expression. GRIM-19 was a target of miR-423-5p, and GRIM-19 was negatively regulated by miR-423-5p in PC3 cells. In addition, miR-423-5p knockdown inhibited the proliferation and promoted the apoptosis of PC3 cells through GRIM-19. In vivo experiments showed that miR-423-5p inhibitor administration reduced tumor volume, down-regulated miR-423-5p and GRIM-19 expressions in PC tissues of nude mice. CONCLUSION Inhibition of miR-423-5p suppressed PC through targeting GRIM-19.
Collapse
Affiliation(s)
- Haili Lin
- Department of Urology, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou 363000, Fujian, China.
| | - Tianqi Lin
- Department of Urology, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou 363000, Fujian, China
| | - Jiangui Lin
- Department of Urology, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou 363000, Fujian, China
| | - Minggen Yang
- Department of Urology, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou 363000, Fujian, China
| | - Zaixiong Shen
- Department of Urology, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou 363000, Fujian, China
| | - Hongjie Liu
- Department of Urology, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou 363000, Fujian, China
| | - Zongkai Zou
- Department of Pathology, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou 363000, Fujian, China
| | - Zhouda Zheng
- Department of Urology, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou 363000, Fujian, China
| |
Collapse
|
25
|
Ni F, Yan CY, Zhou S, Hui PY, Du YH, Zheng L, Yu J, Hu XJ, Zhang ZG. Repression of GRIM19 expression potentiates cisplatin chemoresistance in advanced bladder cancer cells via disrupting ubiquitination-mediated Bcl-xL degradation. Cancer Chemother Pharmacol 2018; 82:593-605. [PMID: 30032449 DOI: 10.1007/s00280-018-3651-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/18/2018] [Indexed: 01/24/2023]
Abstract
OBJECTIVE The mainstay of treatment for advanced bladder cancer (BC) is cisplatin (CDDP)-based systematic chemotherapy. However, acquired chemoresistance induced by as yet unidentified mechanisms is encountered frequently and often results in treatment failure and disease progression. The present study was designed to elucidate the expression and potential role of the gene associated with retinoid-interferon-induced mortality-19 (GRIM19) in the pathogenesis of CDDP resistance in BC. METHODS RT-qPCR and immunoblotting were employed to evaluate the expression profile of GRIM19 in clinical BC samples and in different BC cells. Using cell viability assay, apoptotic ELISA, xenografts mouse model, and Transwell assay, the effects of GRIM19 inhibition or GRIM19 overexpression on CDDP resistance were determined in different BC cells. Lastly, using co-immunoprecipitation, we provided the molecular evidence for the interaction between GRIM19 and Bcl-xL. RESULTS Expression levels of GRIM19 were significantly down-regulated in recurrent BC specimens, and in experimentally induced CDDP-resistant BC cells. Functionally, overexpression of the exogenous GRIM19 potentiated CDDP sensitivity and suppressed the survival and invasion of BC cells in the presence of CDDP challenge. Mechanistically, the compromised CDDP chemosensitization induced by GRIM19 loss was at least partially attributed to the attenuation of Bcl-xL polyubiquitination and subsequent degradation, because (1) GRIM19 colocalized with Bcl-xL in the mitochondria of BC cells and (2) GRIM19 overexpression promoted the ubiquitination of Bcl-xL, and this event could be effectively reversed by pretreatment with inhibitors of p38-MAPK and JNK pathways, indicating that GRIM19 overexpression-induced Bcl-xL ubiquitination may achieve in a p38/JNK-dependent manner. Using the UMUC-3 cells stably depleted of endogenous GRIM19, we further show that inhibition of Bcl-xL rectified GRIM19 deficiency-caused CDDP resistance in BC cells. In addition, BCL2L1 mRNA levels were negatively correlated with GRIM19 mRNA levels in CDDP-associated clinical BC tissues. CONCLUSIONS Disruption of GRIM19/Bcl-xL is a key mechanism of CDDP resistance in advanced BC. Therapeutically, enhancement of GRIM19 expression or employment of p38/JNK inhibitors may serve as resensitizing therapies for subgroups of CDDP-resistant or refractory BC patients.
Collapse
Affiliation(s)
- Feng Ni
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Chang-You Yan
- Family Planning Service Stations of Health and Family Planning Commission of Chengcheng County, Chengcheng County, Weinan City, 714000, Shaanxi Province, China
| | - Sheng Zhou
- Department of Anorectal Surgery, The 2nd Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Baqiao District, Xi'an, 710038, Shaanxi Province, China.
| | - Peng-Yu Hui
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Yong-Hui Du
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Liang Zheng
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Jin Yu
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Xiao-Jian Hu
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Zhi-Gang Zhang
- Department of Urology, The 2nd Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| |
Collapse
|
26
|
The Oncojanus Paradigm of Respiratory Complex I. Genes (Basel) 2018; 9:genes9050243. [PMID: 29735924 PMCID: PMC5977183 DOI: 10.3390/genes9050243] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 04/09/2018] [Accepted: 05/03/2018] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial respiratory function is now recognized as a pivotal player in all the aspects of cancer biology, from tumorigenesis to aggressiveness and chemotherapy resistance. Among the enzymes that compose the respiratory chain, by contributing to energy production, redox equilibrium and oxidative stress, complex I assumes a central role. Complex I defects may arise from mutations in mitochondrial or nuclear DNA, in both structural genes or assembly factors, from alteration of the expression levels of its subunits, or from drug exposure. Since cancer cells have a high-energy demand and require macromolecules for proliferation, it is not surprising that severe complex I defects, caused either by mutations or treatment with specific inhibitors, prevent tumor progression, while contributing to resistance to certain chemotherapeutic agents. On the other hand, enhanced oxidative stress due to mild complex I dysfunction drives an opposite phenotype, as it stimulates cancer cell proliferation and invasiveness. We here review the current knowledge on the contribution of respiratory complex I to cancer biology, highlighting the double-edged role of this metabolic enzyme in tumor progression, metastasis formation, and response to chemotherapy.
Collapse
|
27
|
GRIM-19 represses the proliferation and invasion of cutaneous squamous cell carcinoma cells associated with downregulation of STAT3 signaling. Biomed Pharmacother 2017; 95:1169-1176. [DOI: 10.1016/j.biopha.2017.09.055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/28/2017] [Accepted: 09/12/2017] [Indexed: 12/28/2022] Open
|
28
|
Electron leak from NDUFA13 within mitochondrial complex I attenuates ischemia-reperfusion injury via dimerized STAT3. Proc Natl Acad Sci U S A 2017; 114:11908-11913. [PMID: 29078279 PMCID: PMC5692532 DOI: 10.1073/pnas.1704723114] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Reactive oxygen species (ROS) generation due to electron leak from the mitochondria may be involved in physiological or pathological processes. NDUFA13 is an accessory subunit of mitochondria complex I with a unique molecular structure and is located close to FeS clusters with low electrochemical potentials. Here, we generated cardiac-specific conditional NDUFA13 heterozygous knockout mice. At the basal state, a moderate down-regulation of NDUFA13 created a leak within complex I, resulting in a mild increase in cytoplasm localized H2O2, but not superoxide. The resultant ROS served as a second messenger and was responsible for the STAT3 dimerization and, hence, the activation of antiapoptotic signaling, which eventually significantly suppressed the superoxide burst and decreased the infarct size during the ischemia-reperfusion process. The causative relationship between specific mitochondrial molecular structure and reactive oxygen species (ROS) generation has attracted much attention. NDUFA13 is a newly identified accessory subunit of mitochondria complex I with a unique molecular structure and a location that is very close to the subunits of complex I of low electrochemical potentials. It has been reported that down-regulated NDUFA13 rendered tumor cells more resistant to apoptosis. Thus, this molecule might provide an ideal opportunity for us to investigate the profile of ROS generation and its role in cell protection against apoptosis. In the present study, we generated cardiac-specific tamoxifen-inducible NDUFA13 knockout mice and demonstrated that cardiac-specific heterozygous knockout (cHet) mice exhibited normal cardiac morphology and function in the basal state but were more resistant to apoptosis when exposed to ischemia-reperfusion (I/R) injury. cHet mice showed a preserved capacity of oxygen consumption rate by complex I and II, which can match the oxygen consumption driven by electron donors of N,N,N′,N′-tetramethyl-p-phenylenediamine (TMPD)+ascorbate. Interestingly, at basal state, cHet mice exhibited a higher H2O2 level in the cytosol, but not in the mitochondria. Importantly, increased H2O2 served as a second messenger and led to the STAT3 dimerization and, hence, activation of antiapoptotic signaling, which eventually significantly suppressed the superoxide burst and decreased the infarct size during the I/R process in cHet mice.
Collapse
|