1
|
Liang S, Zhang G, Zhu H, Wei Y, Sun Y, Lin Z, Ling Q. Heat stress induces pathological and molecular responses in the gills of largemouth bass (Micropterus salmoides) revealed by histology, transcriptomics, and DNA methylomics. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101485. [PMID: 40157043 DOI: 10.1016/j.cbd.2025.101485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025]
Abstract
Largemouth bass (Micropterus salmoides), an important species in both aquatic ecosystems and aquaculture, is increasingly affected by heat extremes. Heat stress poses significant challenges to fish health, particularly affecting key organs like the gills. This study investigated the effects of heat stress (37 °C) on the gills of juvenile largemouth bass, focusing on histopathology and molecular adaptations, including genome-wide gene expression and DNA methylation. After the heat stress experiment, histological analysis of hematoxylin and eosin-stained gill tissues revealed significant alterations, including increased blood cell density, epithelial cell proliferation, and curling of gill lamellae. Ultrastructural analysis via transmission electron microscopy showed irregular cell shapes of the secondary lamellae. TUNEL staining demonstrated a significant increase in apoptotic cells, from 14.53 % in control fish versus 69.58 % in heat-stressed fish, which correlated with upregulation of apoptosis-related genes, such as caspase3 and p53. Transcriptomic analysis using RNA sequencing identified 608 differentially expressed genes. KEGG enrichment analysis revealed significant enrichment of the "protein processing in endoplasmic reticulum" pathway. Additionally, whole-genome bisulfite sequencing and methylomic analysis identified 12,505 differentially methylated regions. GO enrichment analysis of genes containing promoter DMRs identified 44 significantly enriched terms, including "metabolic process," "organic substance metabolic process," "binding" and "protein interaction." Notably, genes associated with apoptosis, such as capn3a, dok4, rassf7a, and tusc2a, exhibited significant changes in both gene expression and DNA methylation in their promoters. These findings provide insights into the molecular responses of fish gills to heat stress and mechanisms of resilience in cultured fish species under changing environmental conditions.
Collapse
Affiliation(s)
- Sicheng Liang
- School of Life Sciences, Suzhou Medical College of Soochow University, No. 199 Renai Road, Suzhou, Jiangsu Province 215123, China
| | - Genrong Zhang
- School of Life Sciences, Suzhou Medical College of Soochow University, No. 199 Renai Road, Suzhou, Jiangsu Province 215123, China
| | - Hao Zhu
- School of Life Sciences, Suzhou Medical College of Soochow University, No. 199 Renai Road, Suzhou, Jiangsu Province 215123, China
| | - Yekai Wei
- School of Life Sciences, Suzhou Medical College of Soochow University, No. 199 Renai Road, Suzhou, Jiangsu Province 215123, China
| | - Yi Sun
- Jiangsu Provincial Key Laboratory of Loach Genetics and Breeding, Suzhou Medical College of Soochow University, No. 199 Renai Road, Suzhou, Jiangsu Province 215123, China
| | - Zijie Lin
- School of Life Sciences, Suzhou Medical College of Soochow University, No. 199 Renai Road, Suzhou, Jiangsu Province 215123, China.
| | - Qufei Ling
- School of Life Sciences, Suzhou Medical College of Soochow University, No. 199 Renai Road, Suzhou, Jiangsu Province 215123, China; Jiangsu Provincial Key Laboratory of Loach Genetics and Breeding, Suzhou Medical College of Soochow University, No. 199 Renai Road, Suzhou, Jiangsu Province 215123, China.
| |
Collapse
|
2
|
Liu G, Gao L, Wang Y, Xie X, Gao X, Wu X. The JNK signaling pathway in intervertebral disc degeneration. Front Cell Dev Biol 2024; 12:1423665. [PMID: 39364138 PMCID: PMC11447294 DOI: 10.3389/fcell.2024.1423665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
Intervertebral disc degeneration (IDD) serves as the underlying pathology for various spinal degenerative conditions and is a primary contributor to low back pain (LBP). Recent studies have revealed a strong correlation between IDD and biological processes such as Programmed Cell Death (PCD), cellular senescence, inflammation, cell proliferation, extracellular matrix (ECM) degradation, and oxidative stress (OS). Of particular interest is the emerging evidence highlighting the significant involvement of the JNK signaling pathway in these fundamental biological processes of IDD. This paper explores the potential mechanisms through the JNK signaling pathway influences IDD in diverse ways. The objective of this article is to offer a fresh perspective and methodology for in-depth investigation into the pathogenesis of IDD by thoroughly examining the interplay between the JNK signaling pathway and IDD. Moreover, this paper summarizes the drugs and natural compounds that alleviate the progression of IDD by regulating the JNK signaling pathway. This paper aims to identify potential therapeutic targets and strategies for IDD treatment, providing valuable insights for clinical application.
Collapse
Affiliation(s)
- Ganggang Liu
- Orthopaedics, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lu Gao
- Orthopaedics, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yuncai Wang
- Orthopaedics, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinsheng Xie
- Orthopaedics, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xuejiao Gao
- Otolaryngology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xingjie Wu
- Orthopaedics, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
3
|
Huang Z, Pan T, Xu L, Shi L, Ma X, Zhou L, Wang L, Wang J, Zhu G, Chen D, Song L, Pan X, Wang X, Li X, Luo Y, Chen Y. FGF4 protects the liver from immune-mediated injury by activating CaMKK β-PINK1 signal pathway to inhibit hepatocellular apoptosis. Acta Pharm Sin B 2024; 14:1605-1623. [PMID: 38572102 PMCID: PMC10985030 DOI: 10.1016/j.apsb.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 04/05/2024] Open
Abstract
Immune-mediated liver injury (ILI) is a condition where an aberrant immune response due to various triggers causes the destruction of hepatocytes. Fibroblast growth factor 4 (FGF4) was recently identified as a hepatoprotective cytokine; however, its role in ILI remains unclear. In patients with autoimmune hepatitis (type of ILI) and mouse models of concanavalin A (ConA)- or S-100-induced ILI, we observed a biphasic pattern in hepatic FGF4 expression, characterized by an initial increase followed by a return to basal levels. Hepatic FGF4 deficiency activated the mitochondria-associated intrinsic apoptotic pathway, aggravating hepatocellular apoptosis. This led to intrahepatic immune hyper-reactivity, inflammation accentuation, and subsequent liver injury in both ILI models. Conversely, administration of recombinant FGF4 reduced hepatocellular apoptosis and rectified immune imbalance, thereby mitigating liver damage. The beneficial effects of FGF4 were mediated by hepatocellular FGF receptor 4, which activated the Ca2+/calmodulin-dependent protein kinasekinase 2 (CaMKKβ) and its downstream phosphatase and tensin homologue-induced putative kinase 1 (PINK1)-dependent B-cell lymphoma 2-like protein 1-isoform L (Bcl-XL) signalling axis in the mitochondria. Hence, FGF4 serves as an early response factor and plays a protective role against ILI, suggesting a therapeutic potential of FGF4 and its analogue for treating clinical immune disorder-related liver injuries.
Collapse
Affiliation(s)
- Zhifeng Huang
- Hepatology Diagnosis and Treatment Center, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Tongtong Pan
- Hepatology Diagnosis and Treatment Center, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Liang Xu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University & Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou 325035, China
| | - Lu Shi
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiong Ma
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Liya Zhou
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Luyao Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jiaojiao Wang
- Hepatology Diagnosis and Treatment Center, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Guoqing Zhu
- Hepatology Diagnosis and Treatment Center, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Dazhi Chen
- Hangzhou Medical College, Hangzhou 311300, China
| | - Lingtao Song
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaomin Pan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaodong Wang
- Hepatology Diagnosis and Treatment Center, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaokun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yongde Luo
- Hepatology Diagnosis and Treatment Center, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yongping Chen
- Hepatology Diagnosis and Treatment Center, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
4
|
Jia F, Sun S, Li J, Wang W, Huang H, Hu X, Pan S, Chen W, Shen L, Yao Y, Zheng S, Chen H, Xia W, Yuan H, Zhou J, Yu X, Zhang T, Zhang B, Huang J, Ni C. Neoadjuvant chemotherapy-induced remodeling of human hormonal receptor-positive breast cancer revealed by single-cell RNA sequencing. Cancer Lett 2024; 585:216656. [PMID: 38266804 DOI: 10.1016/j.canlet.2024.216656] [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: 09/19/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 01/26/2024]
Abstract
Hormone receptor-positive breast cancer (HR+ BC) is known to be relatively insensitive to chemotherapy, and since chemotherapy has remained the major neoadjuvant therapy for HR+ BC, the undetermined mechanism of chemoresistance and how chemotherapy reshapes the immune microenvironment need to be explored by high-throughput technology. By using single-cell RNA sequencing and multiplexed immunofluorescence staining analysis of HR+ BC samples (paired pre- and post-neoadjuvant chemotherapy (NAC)), the levels of previously unrecognized immune cell subsets, including CD8+ T cells with pronounced expression of T-cell development (LMNA) and cytotoxicity (FGFBP2) markers, CD4+ T cells characterized by proliferation marker (ATP1B3) expression and macrophages characterized by CD52 expression, were found to be increased post-NAC, which were predictive of chemosensitivity and their antitumor function was also validated with in vitro experiments. In terms of immune checkpoint expression of CD8+ T cells, we found their changes were inconsistent post-NAC, that LAG3, VSIR were decreased, and PDCD1, HAVCR2, CTLA4, KLRC1 and BTLA were increased. In addition, we have identified novel genomic and transcriptional patterns of chemoresistant cancer cells, both innate and acquired, and have confirmed their prognostic value with TCGA cohorts. By shedding light on the ecosystem of HR+ BC reshaped by chemotherapy, our results uncover valuable candidates for predicting chemosensitivity and overcoming chemoresistance in HR+ BC.
Collapse
Affiliation(s)
- Fang Jia
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Shanshan Sun
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China
| | - Jiaxin Li
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Wenwen Wang
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huanhuan Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Xiaoxiao Hu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng Pan
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wuzhen Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lesang Shen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Yao
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siwei Zheng
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hailong Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjie Xia
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Hongjun Yuan
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jun Zhou
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuyan Yu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Zhang
- Department of Radiotherapy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing Zhang
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Jian Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| | - Chao Ni
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China.
| |
Collapse
|
5
|
Xu Y, Du W, Xiao Y, Gao K, Li J, Li S. A Number of the N-terminal RASSF Family: RASSF7. Anticancer Agents Med Chem 2024; 24:889-895. [PMID: 36200241 DOI: 10.2174/1871520622666220930094149] [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: 04/21/2022] [Revised: 07/25/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022]
Abstract
The Ras association domain family 7 (RASSF7, also named HRC1), a potential tumor-related gene, located on human chromosome 11p15, has been identified as an important member of the N-terminal RASSF family. Whereas, the molecular biological mechanisms of RASSF7 in tumorigenesis remain to be further established. We perform a systematic review of the literature and assessment from PUBMED and MEDLINE databases in this article. RASSF7 plays a significant role in mitosis, microtubule growth, apoptosis, proliferation and differentiation. Many research literature shows that the RASSF7 could promote the occurrence and advance of human tumors by regulating Aurora B, MKK4, MKK7, JNK, YAP, MEK, and ERK, whereas, it might inhibit c-Myc and thus lead to the suppression of tumorigenesis. The pregulation of RASSF7 often occurs in various malignancies such as lung cancer, neuroblastoma, thyroid neoplasm, hepatocellular cancer, breast cancer and gastric cancer. The expression stage of RASSF7 is positively correlated with the tumor TNM stage. In this review, we primarily elaborate on the acknowledged structure and progress in the various biomechanisms and research advances of RASSF7, especially the potential relevant signaling pathways. We hope that RASSF7 , a prospective therapeutic target for human malignancies, could play an available role in future anti-cancer treatment.
Collapse
Affiliation(s)
- Yang Xu
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, People's Republic of China
- Department of Urology, Huzhou Central Hospital, Huzhou, Zhejiang, 313000, People's Republic of China
| | - Wei Du
- Department of Urology, Wanbei Coal-Electricity Group General Hospital, Suzhou 234000, People's Republic of China
| | - Yongshuang Xiao
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, People's Republic of China
| | - Keyu Gao
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, People's Republic of China
| | - Jie Li
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, People's Republic of China
| | - Shuofeng Li
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, People's Republic of China
| |
Collapse
|
6
|
Zheng K, Hao F, Medrano-Garcia S, Chen C, Guo F, Morán-Blanco L, Rodríguez-Perales S, Torres-Ruiz R, Peligros MI, Vaquero J, Bañares R, Gómez Del Moral M, Regueiro JR, Martínez-Naves E, Mohamed MR, Gallego-Durán R, Maya D, Ampuero J, Romero-Gómez M, Gilbert-Ramos A, Guixé-Muntet S, Fernández-Iglesias A, Gracia-Sancho J, Coll M, Graupera I, Ginès P, Ciudin A, Rivera-Esteban J, Pericàs JM, Frutos MD, Ramos Molina B, Herranz JM, Ávila MA, Nevzorova YA, Fernández-Malavé E, Cubero FJ. Neuroblastoma RAS viral oncogene homolog (N-RAS) deficiency aggravates liver injury and fibrosis. Cell Death Dis 2023; 14:514. [PMID: 37563155 PMCID: PMC10415403 DOI: 10.1038/s41419-023-06029-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/18/2023] [Accepted: 07/31/2023] [Indexed: 08/12/2023]
Abstract
Progressive hepatic damage and fibrosis are major features of chronic liver diseases of different etiology, yet the underlying molecular mechanisms remain to be fully defined. N-RAS, a member of the RAS family of small guanine nucleotide-binding proteins also encompassing the highly homologous H-RAS and K-RAS isoforms, was previously reported to modulate cell death and renal fibrosis; however, its role in liver damage and fibrogenesis remains unknown. Here, we approached this question by using N-RAS deficient (N-RAS-/-) mice and two experimental models of liver injury and fibrosis, namely carbon tetrachloride (CCl4) intoxication and bile duct ligation (BDL). In wild-type (N-RAS+/+) mice both hepatotoxic procedures augmented N-RAS expression in the liver. Compared to N-RAS+/+ counterparts, N-RAS-/- mice subjected to either CCl4 or BDL showed exacerbated liver injury and fibrosis, which was associated with enhanced hepatic stellate cell (HSC) activation and leukocyte infiltration in the damaged liver. At the molecular level, after CCl4 or BDL, N-RAS-/- livers exhibited augmented expression of necroptotic death markers along with JNK1/2 hyperactivation. In line with this, N-RAS ablation in a human hepatocytic cell line resulted in enhanced activation of JNK and necroptosis mediators in response to cell death stimuli. Of note, loss of hepatic N-RAS expression was characteristic of chronic liver disease patients with fibrosis. Collectively, our study unveils a novel role for N-RAS as a negative controller of the progression of liver injury and fibrogenesis, by critically downregulating signaling pathways leading to hepatocyte necroptosis. Furthermore, it suggests that N-RAS may be of potential clinical value as prognostic biomarker of progressive fibrotic liver damage, or as a novel therapeutic target for the treatment of chronic liver disease.
Collapse
Affiliation(s)
- Kang Zheng
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of Anesthesiology, Nanjing Pukou District Hospital of Chinese Medicine Central Laboratory affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Fengjie Hao
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Sandra Medrano-Garcia
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Chaobo Chen
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- Department of General Surgery, Wuxi Xishan People's Hospital, Wuxi, China
- Department of General Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Feifei Guo
- Department of Obstetrics and Gynaecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Laura Morán-Blanco
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
| | - Sandra Rodríguez-Perales
- Molecular Cytogenetics and Genome Editing Unit, Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Raúl Torres-Ruiz
- Molecular Cytogenetics and Genome Editing Unit, Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - María Isabel Peligros
- Servicio de Anatomía Patológica Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Javier Vaquero
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Rafael Bañares
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Manuel Gómez Del Moral
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of Cell Biology, Complutense University School of Medicine, Madrid, Spain
| | - José R Regueiro
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Eduardo Martínez-Naves
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | | | - Rocío Gallego-Durán
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Douglas Maya
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Javier Ampuero
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Manuel Romero-Gómez
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Albert Gilbert-Ramos
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
| | - Sergi Guixé-Muntet
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
| | - Anabel Fernández-Iglesias
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
| | - Jordi Gracia-Sancho
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Mar Coll
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Laboratorio de Plasticidad de Células Hepáticas y Reparación de Tejidos, Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Isabel Graupera
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Laboratorio de Plasticidad de Células Hepáticas y Reparación de Tejidos, Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Liver Unit, Hospital Clinic, Barcelona, Spain
| | - Pere Ginès
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Liver Unit, Hospital Clinic, Barcelona, Spain
| | - Andreea Ciudin
- Endocrinology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute for Research (VHIR), Barcelona, Spain
| | - Jesús Rivera-Esteban
- Liver Unit, Internal Medicine Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute for Research (VHIR), Barcelona, Spain
| | - Juan M Pericàs
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Unit, Internal Medicine Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute for Research (VHIR), Barcelona, Spain
| | - María Dolores Frutos
- Department of General and Digestive System Surgery, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Bruno Ramos Molina
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Laboratorio de Obesidad y Metabolismo, Instituto de Investigación Biomédica de Murcia (IMIB-Arrixaca), Murcia, Spain
| | - José María Herranz
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Hepatology Programme, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Matías A Ávila
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Hepatology Programme, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Yulia A Nevzorova
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Edgar Fernández-Malavé
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain.
| |
Collapse
|
7
|
Hussey MR, Enquobahrie DA, Loftus CT, MacDonald JW, Bammler TK, Paquette AG, Marsit CJ, Szpiro AA, Kaufman JD, LeWinn KZ, Bush NR, Tylavsky F, Zhao Q, Karr CJ, Sathyanarayana S. Associations of prenatal exposure to NO 2 and near roadway residence with placental gene expression. Placenta 2023; 138:75-82. [PMID: 37216796 PMCID: PMC10349584 DOI: 10.1016/j.placenta.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/03/2023] [Accepted: 05/03/2023] [Indexed: 05/24/2023]
Abstract
INTRODUCTION Traffic-related air pollution (TRAP), a common exposure, potentially impacts pregnancy through altered placental function. We investigated associations between prenatal TRAP exposure and placental gene expression. METHODS Whole transcriptome sequencing was performed on placental samples from CANDLE (Memphis, TN) (n = 776) and GAPPS (Seattle and Yakima, WA) (n = 205), cohorts of the ECHO-PATHWAYS Consortium. Residential NO2 exposures were computed via spatiotemporal models for full-pregnancy, each trimester, and the first/last months of pregnancy. Individual cohort-specific, covariate-adjusted linear models were fit for 10,855 genes and respective exposures (NO2 or roadway proximity [≤150 m]). Infant-sex/exposure interactions on placental gene expression were tested with interaction terms in separate models. Significance was based on false discovery rate (FDR<0.10). RESULTS In GAPPS, final-month NO2 exposure was positively associated with MAP1LC3C expression (FDR p-value = 0.094). Infant-sex interacted with second-trimester NO2 on STRIP2 expression (FDR interaction p-value = 0.011, inverse and positive associations among male and female infants, respectively) and roadway proximity on CEBPA expression (FDR interaction p-value = 0.045, inverse among females). In CANDLE, infant-sex interacted with first-trimester and full-pregnancy NO2 on RASSF7 expression (FDR interaction p-values = 0.067 and 0.013, respectively, positive among male infants and inverse among female infants). DISCUSSION Overall, pregnancy NO2 exposure and placental gene expression associations were primarily null, with exception of final month NO2 exposure and placental MAP1LC3C association. We found several interactions of infant sex and TRAP exposures on placental expression of STRIP2, CEBPA, and RASSF7. These highlighted genes suggest influence of TRAP on placental cell proliferation, autophagy, and growth, though additional replication and functional studies are required for validation.
Collapse
Affiliation(s)
- Michael R Hussey
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA.
| | - Daniel A Enquobahrie
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA; Department of Health Systems and Population Health, School of Public Health, University of Washington, Seattle, WA, USA
| | - Christine T Loftus
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA
| | - Alison G Paquette
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Carmen J Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Adam A Szpiro
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Joel D Kaufman
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA; Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA
| | - Kaja Z LeWinn
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Nicole R Bush
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, School of Medicine, University of California, San Francisco, San, Francisco, CA, USA
| | - Frances Tylavsky
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Qi Zhao
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Catherine J Karr
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA; Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA; Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, USA
| | - Sheela Sathyanarayana
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA; Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA; Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| |
Collapse
|
8
|
Zhu P, Zheng P, Kong X, Wang S, Cao M, Zhao C. Rassf7a promotes spinal cord regeneration and controls spindle orientation in neural progenitor cells. EMBO Rep 2023; 24:e54984. [PMID: 36408859 PMCID: PMC9827555 DOI: 10.15252/embr.202254984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/22/2022] Open
Abstract
Spinal cord injury (SCI) can cause long-lasting disability in mammals due to the lack of axonal regrowth together with the inability to reinitiate spinal neurogenesis at the injury site. Deciphering the mechanisms that regulate the proliferation and differentiation of neural progenitor cells is critical for understanding spinal neurogenesis after injury. Compared with mammals, zebrafish show a remarkable capability of spinal cord regeneration. Here, we show that Rassf7a, a member of the Ras-association domain family, promotes spinal cord regeneration after injury. Zebrafish larvae harboring a rassf7a mutation show spinal cord regeneration and spinal neurogenesis defects. Live imaging shows abnormal asymmetric neurogenic divisions and spindle orientation defects in mutant neural progenitor cells. In line with this, the expression of rassf7a is enriched in neural progenitor cells. Subcellular analysis shows that Rassf7a localizes to the centrosome and is essential for cell cycle progression. Our data indicate a role for Rassf7a in modulating spindle orientation and the proliferation of neural progenitor cells after spinal cord injury.
Collapse
Affiliation(s)
- Panpan Zhu
- Institute of Evolution and Marine BiodiversityOcean University of ChinaQingdaoChina
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
- Sars‐Fang Centre, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life SciencesOcean University of ChinaQingdaoChina
| | - Pengfei Zheng
- Institute of Evolution and Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Xinlong Kong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of PathophysiologyShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuo Wang
- Institute of Evolution and Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Muqing Cao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of PathophysiologyShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chengtian Zhao
- Institute of Evolution and Marine BiodiversityOcean University of ChinaQingdaoChina
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
- Sars‐Fang Centre, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life SciencesOcean University of ChinaQingdaoChina
| |
Collapse
|
9
|
Caliz AD, Vertii A, Fisch V, Yoon S, Yoo HJ, Keaney JF, Kant S. Mitogen-activated protein kinase kinase 7 in inflammatory, cancer, and neurological diseases. Front Cell Dev Biol 2022; 10:979673. [PMID: 36340039 PMCID: PMC9630596 DOI: 10.3389/fcell.2022.979673] [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: 06/27/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Stress-activated mitogen-activated protein kinase kinase 7 (MKK7) is a member of the dual-specificity mitogen-activated protein kinase family. In the human body, MKK7 controls essential physiological processes, including but not limited to proliferation and differentiation in multiple tissues and organs. MKK7, along with the MKK4 pathway, has been implicated in stress-activated activities and biological events that are mediated by c-Jun N-terminal kinase (JNK) signaling. Although numerous studies have been performed to identify the role of JNK in multiple biological processes, there are limited publications that focus on dissecting the independent role of MKK7. Recent research findings have spurred testing via in vivo genetically deficient models, uncovering previously undocumented JNK-independent functions of MKK7. Here we discuss both JNK-dependent and-independent functions of MKK7 in vivo. This review summarizes the role of MKK7 in inflammation, cytokine production, cancer, and neurological diseases.
Collapse
Affiliation(s)
- Amada D Caliz
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Anastassiia Vertii
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Vijay Fisch
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Soonsang Yoon
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Hyung-Jin Yoo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - John F Keaney
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Shashi Kant
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Chen W, Zhang J, Fu H, Hou X, Su Q, He Y, Yang D. KLF5 Is Activated by Gene Amplification in Gastric Cancer and Is Essential for Gastric Cell Proliferation. Cells 2021; 10:cells10051002. [PMID: 33923166 PMCID: PMC8152363 DOI: 10.3390/cells10051002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/12/2021] [Accepted: 04/21/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is the third leading cause of cancer death worldwide. In this study, we tried to clarify the function of KLF5 in gastric cancer. Copy number variation (CNV) and the expression of KLF5 were interrogated in public datasets. The clinical significance of KLF5 amplification and gene expression in gastric cancer were evaluated. The function of KLF5 in cell proliferation was studied in gastric cancer cell lines and organoids. We found that KLF5 amplification mainly occurred in the chromosome instable tumors (CIN) and was significantly associated with TP53 mutation. In addition, higher KLF5 expression correlated with more locally invasive gastric cancer and higher T stage. Next, a KLF5 gene expression signature was curated. The genes in the signature were involved in cell development, cell cycle regulation, cell death, suggesting potential roles played by KLF5. Functional studies using siRNAs revealed that KLF5 was essential for the proliferation of gastric cancer cells. Finally, using gastric organoid models, we revealed that the proliferation of organoids was significantly inhibited after the down regulation of KLF5. Our study revealed that KLF5 was amplified and over-expressed in gastric cancer, and it may play an oncogene-like role in gastric cancer by supporting cell proliferation.
Collapse
Affiliation(s)
- Wei Chen
- Center for Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; (W.C.); (J.Z.); (H.F.); (X.H.)
- Department of Pathology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Jian Zhang
- Center for Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; (W.C.); (J.Z.); (H.F.); (X.H.)
| | - Huafeng Fu
- Center for Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; (W.C.); (J.Z.); (H.F.); (X.H.)
- Laboratory of Surgical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xun Hou
- Center for Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; (W.C.); (J.Z.); (H.F.); (X.H.)
| | - Qiao Su
- Animal Experiment Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
- Correspondence: (Q.S.); (Y.H.); (D.Y.)
| | - Yulong He
- Center for Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; (W.C.); (J.Z.); (H.F.); (X.H.)
- Digestive Medicine Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
- Correspondence: (Q.S.); (Y.H.); (D.Y.)
| | - Dongjie Yang
- Center for Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; (W.C.); (J.Z.); (H.F.); (X.H.)
- Correspondence: (Q.S.); (Y.H.); (D.Y.)
| |
Collapse
|
11
|
Dhanaraman T, Singh S, Killoran RC, Singh A, Xu X, Shifman JM, Smith MJ. RASSF effectors couple diverse RAS subfamily GTPases to the Hippo pathway. Sci Signal 2020; 13:13/653/eabb4778. [PMID: 33051258 DOI: 10.1126/scisignal.abb4778] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Small guanosine triphosphatases (GTPases) of the RAS superfamily signal by directly binding to multiple downstream effector proteins. Effectors are defined by a folded RAS-association (RA) domain that binds exclusively to GTP-loaded (activated) RAS, but the binding specificities of most RA domains toward more than 160 RAS superfamily GTPases have not been characterized. Ten RA domain family (RASSF) proteins comprise the largest group of related effectors and are proposed to couple RAS to the proapoptotic Hippo pathway. Here, we showed that RASSF1-6 formed complexes with the Hippo kinase ortholog MST1, whereas RASSF7-10 formed oligomers with the p53-regulating effectors ASPP1 and ASPP2. Moreover, only RASSF5 bound directly to activated HRAS and KRAS, and RASSFs did not augment apoptotic induction downstream of RAS oncoproteins. Structural modeling revealed that expansion of the RASSF effector family in vertebrates included amino acid substitutions to key residues that direct GTPase-binding specificity. We demonstrated that the tumor suppressor RASSF1A formed complexes with the RAS-related GTPases GEM, REM1, REM2, and the enigmatic RASL12. Furthermore, interactions between RASSFs and RAS GTPases blocked YAP1 nuclear localization. Thus, these simple scaffolds link the activation of diverse RAS family small G proteins to Hippo or p53 regulation.
Collapse
Affiliation(s)
- Thillaivillalan Dhanaraman
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Swati Singh
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Ryan C Killoran
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Anamika Singh
- Hebrew University of Jerusalem, Department of Biological Chemistry, Jerusalem 9190401, Israel
| | - Xingjian Xu
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Julia M Shifman
- Hebrew University of Jerusalem, Department of Biological Chemistry, Jerusalem 9190401, Israel
| | - Matthew J Smith
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec H3T 1J4, Canada. .,Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| |
Collapse
|
12
|
Subramanian DN, Zethoven M, McInerny S, Morgan JA, Rowley SM, Lee JEA, Li N, Gorringe KL, James PA, Campbell IG. Exome sequencing of familial high-grade serous ovarian carcinoma reveals heterogeneity for rare candidate susceptibility genes. Nat Commun 2020; 11:1640. [PMID: 32242007 PMCID: PMC7118163 DOI: 10.1038/s41467-020-15461-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 03/12/2020] [Indexed: 01/31/2023] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) has a significant hereditary component, approximately half of which cannot be explained by known genes. To discover genes, we analyse germline exome sequencing data from 516 BRCA1/2-negative women with HGSOC, focusing on genes enriched with rare, protein-coding loss-of-function (LoF) variants. Overall, there is a significant enrichment of rare protein-coding LoF variants in the cases (p < 0.0001, chi-squared test). Only thirty-four (6.6%) have a pathogenic variant in a known or proposed predisposition gene. Few genes have LoF mutations in more than four individuals and the majority are detected in one individual only. Forty-three highly-ranked genes are identified with three or more LoF variants that are enriched by three-fold or more compared to GnomAD. These genes represent diverse functional pathways with relatively few involved in DNA repair, suggesting that much of the remaining heritability is explained by previously under-explored genes and pathways.
Collapse
Affiliation(s)
- Deepak N Subramanian
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Magnus Zethoven
- Bioinformatics Core Facility, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Simone McInerny
- The Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, 3000, Australia
| | - James A Morgan
- The Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, 3000, Australia
| | - Simone M Rowley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Jue Er Amanda Lee
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Na Li
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kylie L Gorringe
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Paul A James
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- The Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, 3000, Australia
| | - Ian G Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
13
|
Park JG, Aziz N, Cho JY. MKK7, the essential regulator of JNK signaling involved in cancer cell survival: a newly emerging anticancer therapeutic target. Ther Adv Med Oncol 2019; 11:1758835919875574. [PMID: 31579105 PMCID: PMC6759727 DOI: 10.1177/1758835919875574] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 08/19/2019] [Indexed: 01/02/2023] Open
Abstract
One of the mitogen-activated protein kinases (MAPKs), c-Jun NH2-terminal protein kinase (JNK) plays an important role in regulating cell fate, such as proliferation, differentiation, development, transformation, and apoptosis. Its activity is induced through the interaction of MAPK kinase kinases (MAP3Ks), MAPK kinases (MAP2Ks), and various scaffolding proteins. Because of the importance of the JNK cascade to intracellular bioactivity, many studies have been conducted to reveal its precise intracellular functions and mechanisms, but its regulatory mechanisms remain elusive. In this review, we discuss the molecular characterization, activation process, and physiological functions of mitogen-activated protein kinase kinase 7 (MKK7), the MAP2K that most specifically controls the activity of JNK. Understanding the role of MKK7/JNK signaling in physiological conditions could spark new hypotheses for targeted anticancer therapies.
Collapse
Affiliation(s)
- Jae Gwang Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, Republic of Korea
| |
Collapse
|
14
|
Segal T, Salmon-Divon M, Gerlitz G. The Heterochromatin Landscape in Migrating Cells and the Importance of H3K27me3 for Associated Transcriptome Alterations. Cells 2018; 7:E205. [PMID: 30423977 PMCID: PMC6262444 DOI: 10.3390/cells7110205] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 01/01/2023] Open
Abstract
H3K9me3, H3K27me3, and H4K20me1 are epigenetic markers associated with chromatin condensation and transcriptional repression. Previously, we found that migration of melanoma cells is associated with and dependent on global chromatin condensation that includes a global increase in these markers. Taken together with more recent reports by others suggests it is a general signature of migrating cells. Here, to learn about the function of these markers in migrating cells, we mapped them by ChIP-seq analysis. This analysis revealed that induction of migration leads to expansion of these markers along the genome and to an increased overlapping between them. Significantly, induction of migration led to a higher increase in H3K9me3 and H4K20me1 signals at repetitive elements than at protein-coding genes, while an opposite pattern was found for H3K27me3. Transcriptome analysis revealed 182 altered genes following induction of migration, of which 33% are dependent on H3K27me3 for these changes. H3K27me3 was also required to prevent changes in the expression of 501 other genes upon induction of migration. Taken together, our results suggest that heterochromatinization in migrating cells is global and not restricted to specific genomic loci and that H3K27me3 is a key component in executing a migration-specific transcriptional plan.
Collapse
Affiliation(s)
- Tamar Segal
- Department of Molecular Biology, Faculty of Life Sciences and Ariel Center for Applied Cancer Research, Ariel University, Ariel 40700, Israel.
| | - Mali Salmon-Divon
- Department of Molecular Biology, Faculty of Life Sciences and Ariel Center for Applied Cancer Research, Ariel University, Ariel 40700, Israel.
| | - Gabi Gerlitz
- Department of Molecular Biology, Faculty of Life Sciences and Ariel Center for Applied Cancer Research, Ariel University, Ariel 40700, Israel.
| |
Collapse
|
15
|
Kumaraswamy A, Mamidi A, Desai P, Sivagnanam A, Perumalsamy LR, Ramakrishnan C, Gromiha M, Rajalingam K, Mahalingam S. The non-enzymatic RAS effector RASSF7 inhibits oncogenic c-Myc function. J Biol Chem 2018; 293:15691-15705. [PMID: 30139745 DOI: 10.1074/jbc.ra118.004452] [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: 06/14/2018] [Revised: 08/12/2018] [Indexed: 11/06/2022] Open
Abstract
c-Myc is a proto-oncogene controlling expression of multiple genes involved in cell growth and differentiation. Although the functional role of c-Myc as a transcriptional regulator has been intensively studied, targeting this protein in cancer remains a challenge. Here, we report a trimodal regulation of c-Myc function by the Ras effector, Ras-association domain family member 7 (RASSF7), a nonenzymatic protein modulating protein-protein interactions to regulate cell proliferation. Using HEK293T and HeLa cell lines, we provide evidence that RASSF7 destabilizes the c-Myc protein by promoting Cullin4B-mediated polyubiquitination and degradation. Furthermore, RASSF7 competed with MYC-associated factor X (MAX) in the formation of a heterodimeric complex with c-Myc and attenuated its occupancy on target gene promoters to regulate transcription. Consequently, RASSF7 inhibited c-Myc-mediated oncogenic transformation, and an inverse correlation between the expression levels of the RASSF7 and c-Myc genes was evident in human cancers. Furthermore, we found that RASSF7 interacts with c-Myc via its RA and leucine zipper (LZ) domains and LZ domain peptide is sufficient to inhibit c-Myc function, suggesting that this peptide might be used to target oncogenic c-Myc. These results unveil that RASSF7 and c-Myc are functionally linked in the control of tumorigenesis and open up potential therapeutic avenues for targeting the "undruggable" c-Myc protein in a subset of human cancers.
Collapse
Affiliation(s)
- Anbarasu Kumaraswamy
- From the National Cancer Tissue Biobank, Laboratory of Molecular Cell Biology and
| | - Anitha Mamidi
- From the National Cancer Tissue Biobank, Laboratory of Molecular Cell Biology and
| | - Pavitra Desai
- From the National Cancer Tissue Biobank, Laboratory of Molecular Cell Biology and
| | - Ananthi Sivagnanam
- From the National Cancer Tissue Biobank, Laboratory of Molecular Cell Biology and
| | | | - Chandrasekaran Ramakrishnan
- Protein Bioinformatics Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai 600036, India and
| | - Michael Gromiha
- Protein Bioinformatics Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai 600036, India and
| | - Krishnaraj Rajalingam
- the MSU-FZI, Institute of Immunology, University Medical Center Mainz, JGU, 55131 Mainz, Germany
| | | |
Collapse
|
16
|
Zheng X, Dong Q, Zhang X, Han Q, Han X, Han Y, Wu J, Rong X, Wang E. The coiled-coil domain of oncogene RASSF 7 inhibits hippo signaling and promotes non-small cell lung cancer. Oncotarget 2017; 8:78734-78748. [PMID: 29108261 PMCID: PMC5667994 DOI: 10.18632/oncotarget.20223] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 07/12/2017] [Indexed: 11/25/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, and despite recent improvements in treatment patient prognosis remains dismal. In this study, we examined the role of N-terminal Ras-association domain family 7 (RASSF7) in human non-small cell lung cancer (NSCLC). We found that RASSF7 was overexpressed NSCLC tissues, which correlated with advanced TNM stage, positive lymph node metastasis, and poor prognosis. This RASSF7 overexpression promoted lung cancer cell proliferation, migration, and invasion. We also found that RASSF7 interacted with mammalian Ste20-like kinase 1(MST1) through its C-terminal coiled-coil domain to inhibit MST1 phosphorylation as well as the phosphorylation of large tumor suppressor kinase 1(LATS1) and yes-associated protein (YAP), while promoting the nuclear translocation of YAP. In addition, RASSF7 overexpression inhibited the Hippo signaling pathway both in vitro and vivo and promoted the expression of proteins associated with proliferation and invasion, such as connective tissue growth factor. These results suggest that targeting RASSF7 could be exploited for therapeutic benefit in the treatment of NSCLC.
Collapse
Affiliation(s)
- Xiaoying Zheng
- Department of Pathology, College of Basic Medical Science and First Affiliated Hospital, China Medical University, Shenyang, China.,Department of Electron Microscopy, Basic Medical College, Chengde Medical College, Chengde, China
| | - Qianze Dong
- Department of Pathology, College of Basic Medical Science and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiupeng Zhang
- Department of Pathology, College of Basic Medical Science and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Qiang Han
- Department of Pathology, College of Basic Medical Science and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xu Han
- Department of Pathology, College of Basic Medical Science and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yong Han
- Department of Pathology, College of Basic Medical Science and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jingjing Wu
- Department of Pathology, College of Basic Medical Science and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xuezhu Rong
- Department of Pathology, College of Basic Medical Science and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Enhua Wang
- Department of Pathology, College of Basic Medical Science and First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
17
|
Xu S, Dong L, Shi Y, Chen L, Yuan P, Wang S, Li Z, Sun Y, Han S, Yin J, Peng B, He X, Liu W. The Novel Landscape of Long Non-Coding RNAs in Response to Human Foamy Virus Infection Characterized by RNA-Seq. AIDS Res Hum Retroviruses 2017; 33:452-464. [PMID: 27750433 DOI: 10.1089/aid.2016.0156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human foamy virus (HFV) is a complex and unique retrovirus with the longest genomes among retroviruses that are used as vectors for gene therapy. Long non-coding RNAs (lncRNAs) are regarded as key regulators that are involved in diverse biological processes during viral infection. However, the role of lncRNAs in HFV infection remains unknown. In this study, we utilized next-generation sequencing to first characterize lncRNAs in 293T cells after HFV infection, evaluating length distribution, exon number distribution, volcano picture, and lncRNA class distribution. We identified 11,336 lncRNAs (4,729 upregulated lncRNAs and 6,588 downregulated lncRNAs) and 61,367 mRNAs (30,133 upregulated mRNAs and 31,220 downregulated mRNAs), which were differentially expressed in the HFV-infected 293T cells. Subsequently, six differentially expressed lncRNAs characterized using RNA-seq were confirmed by quantitative real-time polymerase chain reaction assays. Interestingly, Gene Ontology (GO)/Gene Ontology Tree Machine (GOTM) and Kyoto Encyclopedia of Gene and Genomes (KEGG) pathway analyses indicated that positive regulation of interleukin 8 (IL8) production and cytokine-cytokine receptor interaction might be involved in the functional enrichment of lncRNAs. Moreover, cis-acting and trans-acting regulatory networks show that NR_028036 may target the fas gene in a cis-acting manner and that ENST00000354838 may target the IL18 gene in a trans-acting manner. Overall, these results not only provide novel insights into the relationship between HFV and lncRNAs in the host response to infection but also have implications for the future wider application of HFV as a vector.
Collapse
Affiliation(s)
- Shanshan Xu
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Lanlan Dong
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Wuhan General Hospital, Guangzhou Military Command, Wuhan, China
| | - Yingying Shi
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Liujun Chen
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Peipei Yuan
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Shuang Wang
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zhi Li
- College of Life Sciences, Shanxi Normal University, Xi'an, Shanxi, China
| | - Yan Sun
- College of Life Sciences, Shanxi Normal University, Xi'an, Shanxi, China
| | - Song Han
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Jun Yin
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Biwen Peng
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Xiaohua He
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Wanhong Liu
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
18
|
Wang T, Feugang JM, Crenshaw MA, Regmi N, Blanton JR, Liao SF. A Systems Biology Approach Using Transcriptomic Data Reveals Genes and Pathways in Porcine Skeletal Muscle Affected by Dietary Lysine. Int J Mol Sci 2017; 18:ijms18040885. [PMID: 28430144 PMCID: PMC5412465 DOI: 10.3390/ijms18040885] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/08/2017] [Accepted: 04/18/2017] [Indexed: 11/16/2022] Open
Abstract
Nine crossbred finishing barrows (body weight 94.4 ± 6.7 kg) randomly assigned to three dietary treatments were used to investigate the effects of dietary lysine on muscle growth related metabolic and signaling pathways. Muscle samples were collected from the longissimus dorsi of individual pigs after feeding the lysine-deficient (4.30 g/kg), lysine-adequate (7.10 g/kg), or lysine-excess (9.80 g/kg) diet for five weeks, and the total RNA was extracted afterwards. Affymetrix Porcine Gene 1.0 ST Array was used to quantify the expression levels of 19,211 genes. Statistical ANOVA analysis of the microarray data showed that 674 transcripts were differentially expressed (at p ≤ 0.05 level); 60 out of 131 transcripts (at p ≤ 0.01 level) were annotated in the NetAffx database. Ingenuity pathway analysis showed that dietary lysine deficiency may lead to: (1) increased muscle protein degradation via the ubiquitination pathway as indicated by the up-regulated DNAJA1, HSP90AB1 and UBE2B mRNA; (2) reduced muscle protein synthesis via the up-regulated RND3 and ZIC1 mRNA; (3) increased serine and glycine synthesis via the up-regulated PHGDH and PSPH mRNA; and (4) increased lipid accumulation via the up-regulated ME1, SCD, and CIDEC mRNA. Dietary lysine excess may lead to: (1) decreased muscle protein degradation via the down-regulated DNAJA1, HSP90AA1, HSPH1, and UBE2D3 mRNA; and (2) reduced lipid biosynthesis via the down-regulated CFD and ME1 mRNA. Collectively, dietary lysine may function as a signaling molecule to regulate protein turnover and lipid metabolism in the skeletal muscle of finishing pigs.
Collapse
Affiliation(s)
- Taiji Wang
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA.
| | - Jean M Feugang
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA.
| | - Mark A Crenshaw
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA.
| | - Naresh Regmi
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA.
| | - John R Blanton
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA.
| | - Shengfa F Liao
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA.
| |
Collapse
|
19
|
Guo M, Wei J, Zhou Y, Qin Q. MKK7 confers different activities to viral infection of Singapore grouper iridovirus (SGIV) and nervous necrosis virus (NNV) in grouper. FISH & SHELLFISH IMMUNOLOGY 2016; 57:419-427. [PMID: 27601297 DOI: 10.1016/j.fsi.2016.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/13/2016] [Accepted: 09/02/2016] [Indexed: 06/06/2023]
Abstract
Mitogen-activated protein kinase 7 (MKK7) is one of the major stress-activated protein kinase (SAPK)-activating kinases in response to environmental or physiological stimuli. Here a MKK7 named as Ec-MKK7 was identified from orange-spotted grouper, Epinephelus coioides. The full-length cDNA of Ec-MKK7 was 1853 bp, with an open reading frame (ORF) of 1272 bp encoding a putative protein of 423 amino acids. A characteristic S-K-A-K-T motif was contained in the domain of dual-specificity protein kinase, mitogen-activated protein kinase kinase 7 (PKc_MKK7). Intracellular localization showed that Ec-MKK7 was localized in both the cytoplasm and the nucleus of grouper spleen (GS) and/or grouper brain (EAGB) cells. Moreover, Ec-MKK7 was universally expressed in all examined tissues and showed expression modulation to challenges of lipopolysacchride (LPS), Singapore grouper iridovirus (SGIV) and polyriboinosinic polyribocytidylic acid (poly I:C) in vivo. A gene targeting strategy over-expressing Ec-MKK7 was performed to examine the activities of MKK7 to viral infection in vitro. Our data showed that Ec-MKK7 was involved in the evasion and replication of SGIV but played an antiviral role to the infection of nervous necrosis virus (NNV). All results demonstrated that Ec-MKK7 could play important roles in grouper innate immunity and show distinct functions on virus infection.
Collapse
Affiliation(s)
- Minglan Guo
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, PR China; Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, PR China
| | - Jingguang Wei
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, PR China; Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, PR China
| | - Yongcan Zhou
- State Key Laboratory of Marine Resource Utilization in South China Sea, College of Marine Science, Hainan University, Haikou, 570228, PR China.
| | - Qiwei Qin
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, PR China; College of Marine Sciences, South China Agricultural University, Guangzhou 510301, PR China.
| |
Collapse
|
20
|
Sarkar H, Arya S, Rai U, Majumdar SS. A Study of Differential Expression of Testicular Genes in Various Reproductive Phases of Hemidactylus flaviviridis (Wall Lizard) to Derive Their Association with Onset of Spermatogenesis and Its Relevance to Mammals. PLoS One 2016; 11:e0151150. [PMID: 26963275 PMCID: PMC4795608 DOI: 10.1371/journal.pone.0151150] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 02/24/2016] [Indexed: 12/19/2022] Open
Abstract
Testis of Hemidactylus flaviviridis, commonly known as Indian wall lizard, displays a lack of cellular and metabolic activity in regressed phase of testis during non-breeding season of the year. Retracted Sertoli cells (Sc), fibroid myoid cells and pre-meiotic resting spermatogonia are observed in such testis. This situation is akin to certain forms of infertility in men where hormone supplementation fails to generate sperm despite the presence of Sc and germ cells (Gc) in testis. In testis of lizard, spermatogenesis is reinitiated upon increased level of hormones during appropriate season (phase of recrudescence). Study of genes associated with generation of sperm, from regressed adult testis in lizard, may provide valuable information for understanding certain forms of male idiopathic infertility. Subtractive hybridization using testicular RNA obtained from the regressed and active phases of lizard reproductive cycle led to identify eight partial mRNA sequences that showed sequence homology with mice genes. We further evaluated the gene expression prolife by real-time PCR in three different reproductive phases of H. flaviviridis: regressed (pre-meiotic), recrudescent (meiotic) and active (post meiotic), for comparison with the corresponding testicular phases found in testis of 5 days (pre-meiotic), 20 days (meiotic) and 60 days (post-meiotic) old mouse. This is the first report where genes associated with progression of spermatogenesis during active phase, which follows a regressed state of adult testis, were identified in lizard and found to be conserved in mouse. Six important genes, Hk1, Nme5, Akap4, Arih1, Rassf7 and Tubb4b were found to be strictly associated with active spermatogenesis in both mouse and lizard. Factors interfering with the expression of any of these genes may potentially abrogate the process of spermatogenesis leading to infertility. Such information may shed light on unknown causes of idiopathic male infertility.
Collapse
Affiliation(s)
- Hironmoy Sarkar
- Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Satyapal Arya
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Umesh Rai
- Department of Zoology, University of Delhi, Delhi, 110007, India
- * E-mail: (UR); (SSM)
| | - Subeer S. Majumdar
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India
- * E-mail: (UR); (SSM)
| |
Collapse
|
21
|
Liu ZH, Huo JL, Wu ZG, Sun Z, Bai F, Samartzis D, Gantenbein B, Fan SD, Wang HQ. RASSF7 expression and its regulatory roles on apoptosis in human intervertebral disc degeneration. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:16097-16103. [PMID: 26884887 PMCID: PMC4730100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/17/2015] [Indexed: 06/05/2023]
Abstract
Apoptosis plays an important role in intervertebral disc degeneration (IDD). Overwhelming evidence indicates that RASSF7 is essential for cell growth and apoptosis. Recently, it has been noted that the JNK signaling can be negatively regulated by suppressing phosphorylated-MKK7 activation during pro-apoptosis. We aimed to investigate the RASSF7 expression level in human degenerative nucleus pulposus (NP) cells and non-degenerative NP cells and the link between RASSF7-JNK with NP cells apoptosis. We harvested NP tissues from 20 IDD patients as disease group and 8 cadaveric donors as normal controls. We detected RASSF7 expression by Real-time-PCR and western blotting. Consequently, we found that the expression of RASSF7 was higher in non-degenerative group than in degenerative group (P<0.05). Overexpression of RASSF7 in degenerative NP cells led to decreased apoptosis rate than that in scramble group (P<0.05). Collectively, our findings suggest that RASSF7 plays an important role in human IDD and RASSF7 might be potentially developed as a curative agent.
Collapse
Affiliation(s)
- Zhi-Heng Liu
- Department of Orthopaedics, Xi’an Air Force Hospital, PLA172 Youyi Eastern Road, Xi’an, P. R. China
| | - Jun-Li Huo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University127 Changle Western Road, Xi’an, P. R. China
| | - Zhi-Gang Wu
- Department of Orthopaedics, Lanzhou General Hospital of Lanzhou Military Region, PLALanzhou, P. R. China
| | - Zhen Sun
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University127 Changle Western Road, Xi’an, P. R. China
| | - Feng Bai
- Department of Orthopaedics, Xi’an Air Force Hospital, PLA172 Youyi Eastern Road, Xi’an, P. R. China
| | - Dino Samartzis
- Department of Orthopaedics and Traumatology, The University of Hong KongPokfulam, Hong Kong, SAR China
| | - Benjamin Gantenbein
- Institute for Surgical Technology and Biomechanics, Tissue and Organ Mechanobiology, University of BernBern, Switzerland
| | - Shao-Di Fan
- Department of Orthopaedics, Xi’an Air Force Hospital, PLA172 Youyi Eastern Road, Xi’an, P. R. China
| | - Hai-Qiang Wang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University127 Changle Western Road, Xi’an, P. R. China
| |
Collapse
|
22
|
Gulsen T, Hadjicosti I, Li Y, Zhang X, Whitley PR, Chalmers AD. Truncated RASSF7 promotes centrosomal defects and cell death. Dev Biol 2015; 409:502-17. [PMID: 26569555 DOI: 10.1016/j.ydbio.2015.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 03/31/2015] [Accepted: 11/02/2015] [Indexed: 10/22/2022]
Abstract
RASSF7 protein localises to the centrosome and plays a key role in mitosis. Its expression is also increased in a range of tumour types. However, little is known about the molecular basis of RASSF7's function and it is not clear if it acts as an oncogene in the cancers where its levels are elevated. Here, we carry out the first analysis of the domains of rassf7, focusing on which of them are responsible for its localisation to the centrosome. Constructs were generated to allow the expression of a series of truncated versions of rassf7 and the level of centrosomal localisation shown by each protein quantified. This analysis was carried out in Xenopus embryos which are a tractable system where rassf7 localisation can easily be studied. Our data shows that the coiled-coil domain of rassf7 is required and sufficient to direct its centrosomal localisation. The RA domain did not appear to have a role in mediating localisation. Surprisingly, removal of the extreme C-terminus of the protein caused rassf7 to accumulate at the centrosome and drive centrosome defects, including accumulation of the centrosomal protein γ-tubulin and an amplification of the number of γ-tubulin foci. These effects required the centrosomal localisation mediated by the coiled-coil domain. Later in development cells expressing this truncated rassf7 protein underwent cell death. Finally, analysis of a database of tumour sequences identified a mutation in RASSF7 which would cause a similar C-terminal truncation of the protein. Based on our data this truncated protein might drive centrosomal defects and we propose the hypothesis that truncated RASSF7 could act as an oncogene in a small subset of tumours where it is mutated in this way.
Collapse
Affiliation(s)
- Tulay Gulsen
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Irene Hadjicosti
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Yueshi Li
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Xinyun Zhang
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Paul R Whitley
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Andrew D Chalmers
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom.
| |
Collapse
|
23
|
McLean LS, Watkins CN, Campbell P, Zylstra D, Rowland L, Amis LH, Scott L, Babb CE, Livingston WJ, Darwanto A, Davis WL, Senthil M, Sowers LC, Brantley E. Aryl Hydrocarbon Receptor Ligand 5F 203 Induces Oxidative Stress That Triggers DNA Damage in Human Breast Cancer Cells. Chem Res Toxicol 2015; 28:855-71. [PMID: 25781201 DOI: 10.1021/tx500485v] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Breast tumors often show profound sensitivity to exogenous oxidative stress. Investigational agent 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (5F 203) induces aryl hydrocarbon receptor (AhR)-mediated DNA damage in certain breast cancer cells. Since AhR agonists often elevate intracellular oxidative stress, we hypothesize that 5F 203 increases reactive oxygen species (ROS) to induce DNA damage, which thwarts breast cancer cell growth. We found that 5F 203 induced single-strand break formation. 5F 203 enhanced oxidative DNA damage that was specific to breast cancer cells sensitive to its cytotoxic actions, as it did not increase oxidative DNA damage or ROS formation in nontumorigenic MCF-10A breast epithelial cells. In contrast, AhR agonist and procarcinogen benzo[a]pyrene and its metabolite, 1,6-benzo[a]pyrene quinone, induced oxidative DNA damage and ROS formation, respectively, in MCF-10A cells. In sensitive breast cancer cells, 5F 203 activated ROS-responsive kinases: c-Jun-N-terminal kinase (JNK) and p38 mitogen activated protein kinase (p38). AhR antagonists (alpha-naphthoflavone, CH223191) or antioxidants (N-acetyl-l-cysteine, EUK-134) attenuated 5F 203-mediated JNK and p38 activation, depending on the cell type. Pharmacological inhibition of AhR, JNK, or p38 attenuated 5F 203-mediated increases in intracellular ROS, apoptosis, and single-strand break formation. 5F 203 induced the expression of cytoglobin, an oxidative stress-responsive gene and a putative tumor suppressor, which was diminished with AhR, JNK, or p38 inhibition. Additionally, 5F 203-mediated increases in ROS production and cytoglobin were suppressed in AHR100 cells (AhR ligand-unresponsive MCF-7 breast cancer cells). Our data demonstrate 5F 203 induces ROS-mediated DNA damage at least in part via AhR, JNK, or p38 activation and modulates the expression of oxidative stress-responsive genes such as cytoglobin to confer its anticancer action.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Lawrence C Sowers
- ⊥Department of Pharmacology and Toxicology, University of Texas Medical Branch at Galveston, Galveston, Texas 77555, United States
| | | |
Collapse
|
24
|
Volodko N, Gordon M, Salla M, Ghazaleh HA, Baksh S. RASSF tumor suppressor gene family: Biological functions and regulation. FEBS Lett 2014; 588:2671-84. [DOI: 10.1016/j.febslet.2014.02.041] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 02/25/2014] [Accepted: 02/25/2014] [Indexed: 01/22/2023]
|
25
|
Li Z, Chang X, Dai D, Deng P, Sun Q. RASSF10 is an epigenetically silenced tumor suppressor in gastric cancer. Oncol Rep 2014; 31:1661-8. [PMID: 24573726 DOI: 10.3892/or.2014.3039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 01/14/2014] [Indexed: 11/06/2022] Open
Abstract
To better understand the role of the N-Terminal Ras association domain family (RASSF) genes in the development of gastric cancer, we examined the expression of RASSF7 and RASSF10 and RASSF10 methylation in gastric cancer. We found that RASSF10 expression was lost in six gastric cancer cell lines, and was rescued by a DNA demethylating agent and a histone deacetylase inhibitor. However, RASSF7 expression was strong in four cancer cell lines as well as in 87% of primary gastric cancer tissues. In contrast, RASSF7 expression was moderate in the GES-1 cell line and negative in 33.3% of the corresponding non-cancerous tissues. Analysis of RASSF10 methylation by methylation-specific PCR (MSP) and sequencing revealed that the methylation frequency in primary gastric carcinoma tissues was significantly higher compared to that in adjacent non-carcinoma tissues (61.6 vs. 38.4%; p<0.01). The methylation frequency in the tumor with invasion depth at T3 and T4 was significantly higher compared to that with invasion depth at T1 and T2 (67.1 vs. 37.5%; p<0.05). Hypermethylation of RASSF10 was found in the patients with lymph node metastasis, compared to those with unaffected lymph nodes (68.8 vs. 40.9%; p<0.05). Among the 4 gross types of the Borrmann classification, i.e. EGC, Borrmann Ⅰ, Borrmann Ⅱ, Borrmann Ⅲ and Borrmann Ⅳ, the last one was more frequently methylated (85.7 vs. 56.9%; p<0.05). The present study revealed that RASSF10 is an epigenetically silenced gene involved in tumor invasion and metastasis in gastric cancer, suggesting that the methylation status of RASSF10 may be a useful indicator to predict the malignant degree of gastric cancer.
Collapse
Affiliation(s)
- Zhenhua Li
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Xiaojing Chang
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Dongqiu Dai
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Peng Deng
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Qiang Sun
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, P.R. China
| |
Collapse
|
26
|
Chan JJ, Katan M. PLCɛ and the RASSF family in tumour suppression and other functions. Adv Biol Regul 2013; 53:258-279. [PMID: 23958207 DOI: 10.1016/j.jbior.2013.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 07/15/2013] [Indexed: 06/02/2023]
Abstract
Not all proteins implicated in direct binding to Ras appear to have a positive role in the generation and progression of tumours; examples include Phospholipase C epsilon (PLCɛ) and some members of the Ras-association domain family (RASSF). The RASSF family comprises of ten members, known as RASSF1 to RASSF10. PLCɛ and RASSF members carry a common Ras-association domain (RA) that can potentially bind Ras oncoproteins and mediate Ras-regulated functions. RASSF1 to RASSF6 also share a common SARAH domain that facilitates protein-protein interactions with other SARAH domain proteins. The majority of the family are frequently downregulated by epigenetic silencing in cancers. They are implicated in various important biological processes including apoptosis, microtubule stabilisation and cell cycle regulation. Recent studies have reinforced the tumour suppressive properties of the RASSF family, with new evidence of emerging pathways and novel functions that suggest a wider role for these proteins. This review will first describe an emerging role of PLCɛ in tumour suppression and then focus on and summarise the new findings on the RASSF family in the last five years to consolidate their well-established functions, and highlight the new regulatory roles of specific RASSF members.
Collapse
Affiliation(s)
- Jia Jia Chan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | | |
Collapse
|
27
|
Chan JJ, Flatters D, Rodrigues-Lima F, Yan J, Thalassinos K, Katan M. Comparative analysis of interactions of RASSF1-10. Adv Biol Regul 2013; 53:190-201. [PMID: 23357313 PMCID: PMC4221134 DOI: 10.1016/j.jbior.2012.12.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 12/13/2012] [Indexed: 01/01/2023]
Abstract
Members of the RASSF family (RASSF1-10) have been identified as candidate tumour suppressors that are frequently downregulated by promoter hypermethylation in cancers. These proteins carry a common Ras-association (RA) and SARAH domain (RASSF1-6) that can potentially bind Ras oncoproteins and mediate protein-protein interactions with other SARAH domain proteins. However, there is a notable lack of comparative characterisation of the RASSF family, as well as molecular and structural information that facilitate their tumour suppressive functions. As part of our comparative analysis, we modelled the RA and SARAH domains of the RASSF members based on existing structures and predicted their potential interactions. These in silico predictions were compared to in vitro interaction studies with Ras and MST kinase (a SARAH domain-containing protein). Our data shows a diversity of interaction within the RASSF family RA domain, whereas the SARAH domain-mediated interactions for RASSF1-6 are consistent with the predictions. This suggests that different members, despite shared general architecture, could have distinct functional properties. Additionally, we identify a new interacting partner for MST kinase in the form of RASSF7. Current data supports an interaction model where RASSF serves as an adaptor for the assembly of multiple protein complexes and further functional interactions, involving MST kinases and other SARAH domain proteins, which could be regulated by Ras.
Collapse
Affiliation(s)
- Jia Jia Chan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - Delphine Flatters
- Université Paris Diderot, Sorbonne Paris Cité, Molécules Thérapeutiques in silico, Inserm UMR-S 973, 35 rue Helene Brion, 75013 Paris, France
| | - Fernando Rodrigues-Lima
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS EAC4413, 75013, Paris, France
| | - Jun Yan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - Konstantinos Thalassinos
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - Matilda Katan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
28
|
Growth cone MKK7 mRNA targeting regulates MAP1b-dependent microtubule bundling to control neurite elongation. PLoS Biol 2012; 10:e1001439. [PMID: 23226105 PMCID: PMC3514283 DOI: 10.1371/journal.pbio.1001439] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 10/24/2012] [Indexed: 11/19/2022] Open
Abstract
Local mRNA translation in neurons has been mostly studied during axon guidance and synapse formation but not during initial neurite outgrowth. We performed a genome-wide screen for neurite-enriched mRNAs and identified an mRNA that encodes mitogen-activated protein kinase kinase 7 (MKK7), a MAP kinase kinase (MAPKK) for Jun kinase (JNK). We show that MKK7 mRNA localizes to the growth cone where it has the potential to be translated. MKK7 is then specifically phosphorylated in the neurite shaft, where it is part of a MAP kinase signaling module consisting of dual leucine zipper kinase (DLK), MKK7, and JNK1. This triggers Map1b phosphorylation to regulate microtubule bundling leading to neurite elongation. We propose a model in which MKK7 mRNA localization and translation in the growth cone allows for a mechanism to position JNK signaling in the neurite shaft and to specifically link it to regulation of microtubule bundling. At the same time, this uncouples activated JNK from its functions relevant to nuclear translocation and transcriptional activation.
Collapse
|
29
|
Malla RR, Gopinath S, Alapati K, Gorantla B, Gondi CS, Rao JS. uPAR and cathepsin B inhibition enhanced radiation-induced apoptosis in gliomainitiating cells. Neuro Oncol 2012; 14:745-60. [PMID: 22573309 DOI: 10.1093/neuonc/nos088] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Glioblastomas present as diffuse tumors with invasion into normal brain tissue and frequently recur or progress after radiation as focal masses because of glioma-initiating cells. The role of the urokinase-type plasminogen activator receptor (uPAR) and cathepsin B in stem-like phenotype has been extensively studied in several solid tumors. In the present study, we demonstrated that selection of glioma-initiating cells using CD133 expression leads to a specific enrichment of CD133(+) cells in both U87 and 4910 cells. In addition, CD133(+) cells exhibited a considerable amount of other stem cell markers, such as Nestin and Sox-2. Radiation treatment significantly enhanced uPAR and cathepsin B levels in glioma-initiating cells. To downregulate radiation-induced uPAR and cathepsin B expression, we used a bicistronic shRNA construct that simultaneously targets both uPAR and cathepsin B (pCU). Downregulation of uPAR and cathepsin B using pCU decreased radiation-enhanced uPAR and cathepsin B levels and caused DNA damage-induced apoptosis in glioma cell lines and glioma-initiating cells. The most striking finding of this study is that knockdown of uPAR and cathepsin B inhibited ongoing transcription by suppressing BrUTP incorporation at γH2AX foci. In addition, uPAR and cathepsin B gene silencing inversely regulated survivin and H2AX expression in both glioma cells and glioma-initiating cells. Pretreatment with pCU reduced radiation-enhanced expression of uPAR, cathepsin B, and survivin and enhanced DNA damage in pre-established glioma in nude mice. Taken together, our in vitro and in vivo findings suggest that uPAR and cathepsin B inhibition might serve as an adjunct to radiation therapy to target glioma-initiating cells and, therefore, for the treatment of glioma.
Collapse
Affiliation(s)
- Rama Rao Malla
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, IL 61605, USA
| | | | | | | | | | | |
Collapse
|
30
|
Hata S, Hirayama J, Kajiho H, Nakagawa K, Hata Y, Katada T, Furutani-Seiki M, Nishina H. A novel acetylation cycle of transcription co-activator Yes-associated protein that is downstream of Hippo pathway is triggered in response to SN2 alkylating agents. J Biol Chem 2012; 287:22089-98. [PMID: 22544757 DOI: 10.1074/jbc.m111.334714] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Yes-associated protein (YAP) is a transcriptional co-activator that acts downstream of the Hippo signaling pathway and regulates multiple cellular processes. Although cytoplasmic retention of YAP is known to be mediated by Hippo pathway-dependent phosphorylation, post-translational modifications that regulate YAP in the nucleus remain unclear. Here we report the discovery of a novel cycle of acetylation/deacetylation of nuclear YAP induced in response to S(N)2 alkylating agents. We show that after treatment of cells with the S(N)2 alkylating agent methyl methanesulfonate, YAP phosphorylation mediated by the Hippo pathway is markedly reduced, leading to nuclear translocation of YAP and its acetylation. This YAP acetylation occurs on specific and highly conserved C-terminal lysine residues and is mediated by the nuclear acetyltransferases CBP (CREB binding protein) and p300. Conversely, the nuclear deacetylase SIRT1 is responsible for YAP deacetylation. Intriguingly, we found that YAP acetylation is induced specifically by S(N)2 alkylating agents and not by other DNA-damaging stimuli. These results identify a novel YAP acetylation cycle that occurs in the nucleus downstream of the Hippo pathway. Intriguingly, our findings also indicate that YAP acetylation is involved in responses to a specific type of DNA damage.
Collapse
Affiliation(s)
- Shoji Hata
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
RASSF Signalling and DNA Damage: Monitoring the Integrity of the Genome? Mol Biol Int 2012; 2012:141732. [PMID: 22577550 PMCID: PMC3337673 DOI: 10.1155/2012/141732] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 01/27/2012] [Indexed: 12/14/2022] Open
Abstract
The RASSF family of proteins has been extensively studied in terms of their genetics, structure and function. One of the functions that has been increasingly studied is the role of the RASSF proteins in the DNA damage response. Surprisingly, this research, which encompasses both the classical and N-terminal RASSF proteins, has revealed an involvement of the RASSFs in oncogenic pathways as well as the more familiar tumour suppressor pathways usually associated with the RASSF family members. The most studied protein with respect to DNA damage is RASSF1A, which has been shown, not only to be activated by ATM, a major regulator of the DNA damage response, but also to bind to and activate a number of different pathways which all lead to and feedback from the guardian of the genome, p53. In this review we discuss the latest research linking the RASSF proteins to DNA damage signalling and maintenance of genomic integrity and look at how this knowledge is being utilised in the clinic to enhance the effectiveness of traditional cancer therapies such as radiotherapy.
Collapse
|
32
|
Exploring the function of the JNK (c-Jun N-terminal kinase) signalling pathway in physiological and pathological processes to design novel therapeutic strategies. Biochem Soc Trans 2012; 40:85-9. [DOI: 10.1042/bst20110641] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
JNK (c-Jun N-terminal kinase) is a member of the MAPK (mitogen-activated protein kinase) family that regulates a range of biological processes implicated in tumorigenesis and neurodegenerative disorders. For example, genetic studies have demonstrated that the removal of specific Jnk genes can reduce neuronal death associated with cerebral ischaemia. As such, targeting JNK signalling constitutes an obvious opportunity for therapeutic intervention. However, MAPK inhibitors can display toxic effects. Consequently, dual-specificity MKKs (MAPK kinases) may represent more attractive targets. In particular, evidence that blocking JNK activation by removing MKK4 offers an effective therapy to treat pathological conditions has started to emerge. MKK4 was the first JNK activator identified. The remaining level of JNK activity in cells lacking MKK4 expression led to the discovery of a second activator of JNK, named MKK7. Distinct phenotypic abnormalities associated with the targeted deletion of Mkk4 and Mkk7 in mice have revealed that MKK4 and MKK7 have non-redundant function in vivo. Further insights into the specific functions of the JNK activators in cancer cells and in neurons will be of critical importance to validate MKK4 and MKK7 as promising drug targets.
Collapse
|