1
|
Sant'ana JF, Tureta EF, Rosa RLDA, Calegari-Alves YP, Faustino AM, Marques AL, Bobermin LD, Quincozes-Santos A, Varela APM, Sesterheim P, Berger M, Peña RD, Souza DO, Roehe P, Guimarães JA, Campos AR, Santi L, Beys-DA-Silva WO. Zika virus infection in a non-neural cell host promotes differential expression of proteins associated with neurological conditions. AN ACAD BRAS CIENC 2025; 97:e20240849. [PMID: 40197952 DOI: 10.1590/0001-3765202520240849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/09/2024] [Indexed: 04/10/2025] Open
Abstract
The Zika Virus (ZIKV) is a Flavivirus that caused a recent outbreak worldwide resulting in different neurological outcomes that are still poorly characterized and understood. Concerning this issue, in vitro and in vivo models are being applied to improve the molecular understanding of ZIKV infection. In this work, applying shotgun proteomics we revealed the differential ZIKV infection proteome in Vero cells, a non-neural cell model. A dramatic change resulting from infection was found including the differential expression of several proteins previously associated with brain diseases. The molecular alterations caused by this pathogen were further characterized through bioinformatics such as Gene Ontology and protein-protein interaction network of resulting differential proteome. Our findings identified molecular markers that were differentially expressed during ZIKV infection and had been previously linked to neurological conditions and infections caused by ZIKV and/or SARS-CoV-2. The results presented in this article highlight molecular markers associated with neurological dysfunctions, demonstrating that ZIKV infection can dysregulate neural-specific genes, even in non-neural cells.
Collapse
Affiliation(s)
- Júlia F Sant'ana
- Universidade Federal do Rio Grande do Sul, Faculdade de Farmácia, Av. Ipiranga, 2752, suite 709, 90160-093 Porto Alegre, RS, Brazil
| | - Emanuela Fernanda Tureta
- Universidade Federal do Rio Grande do Sul, Faculdade de Farmácia, Av. Ipiranga, 2752, suite 709, 90160-093 Porto Alegre, RS, Brazil
| | - Rafael L DA Rosa
- Universidade Federal do Rio Grande do Sul, Faculdade de Farmácia, Av. Ipiranga, 2752, suite 709, 90160-093 Porto Alegre, RS, Brazil
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Rua Ramiro Barcelos, 2350, 90035-007 Porto Alegre, RS, Brazil
| | - Yohana P Calegari-Alves
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Rua Ramiro Barcelos, 2350, 90035-007 Porto Alegre, RS, Brazil
| | - Aline M Faustino
- Universidade Federal do Rio Grande do Sul, Faculdade de Farmácia, Av. Ipiranga, 2752, suite 709, 90160-093 Porto Alegre, RS, Brazil
| | - Ana Luiza Marques
- Universidade Federal do Rio Grande do Sul, Faculdade de Farmácia, Av. Ipiranga, 2752, suite 709, 90160-093 Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos, 2600, Suite 625, 90035-003 Porto Alegre, RS, Brazil
| | - André Quincozes-Santos
- Universidade Federal do Rio Grande do Sul, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, RS, Brazil
| | - Ana Paula M Varela
- Instituto de Cardiologia/Fundação Universitária de Cardiologia, Centro de Cardiologia Experimental, Rua Domingos Crescêncio, 132, 90650-090 Porto Alegre, RS, Brazil
| | - Patrícia Sesterheim
- Instituto de Cardiologia/Fundação Universitária de Cardiologia, Centro de Cardiologia Experimental, Rua Domingos Crescêncio, 132, 90650-090 Porto Alegre, RS, Brazil
| | - Markus Berger
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Rua Ramiro Barcelos, 2350, 90035-007 Porto Alegre, RS, Brazil
| | - Ramon D Peña
- Sanford Burnham Prebys Medical Discovery Institute, Proteomics Core, 10901N, Torrey Pines Road, 92037, La Jolla, CA, USA
| | - Diogo O Souza
- Universidade Federal do Rio Grande do Sul, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, RS, Brazil
| | - Paulo Roehe
- Universidade Federal do Rio Grande do Sul, Departamento de Microbiologia, Instituto de Ciências Básicas da Saúde, Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - Jorge A Guimarães
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Rua Ramiro Barcelos, 2350, 90035-007 Porto Alegre, RS, Brazil
| | - Alexandre R Campos
- Sanford Burnham Prebys Medical Discovery Institute, Proteomics Core, 10901N, Torrey Pines Road, 92037, La Jolla, CA, USA
| | - Lucélia Santi
- Universidade Federal do Rio Grande do Sul, Faculdade de Farmácia, Av. Ipiranga, 2752, suite 709, 90160-093 Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Centro de Biotecnologia, Av. Bento Gonçalves, 9500, 91501-970 Porto Alegre, RS, Brazil
| | - Walter Orlando Beys-DA-Silva
- Universidade Federal do Rio Grande do Sul, Faculdade de Farmácia, Av. Ipiranga, 2752, suite 709, 90160-093 Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Centro de Biotecnologia, Av. Bento Gonçalves, 9500, 91501-970 Porto Alegre, RS, Brazil
| |
Collapse
|
2
|
Chang X, Ji C, Zhang T, Huang H. Prenatal to preimplantation genetic diagnosis of a novel compound heterozygous mutation in HSPA9 associated with Even-Plus syndrome. Clin Chim Acta 2024; 555:117803. [PMID: 38281662 DOI: 10.1016/j.cca.2024.117803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 01/30/2024]
Abstract
BACKGROUND Heat shock protein family A member 9 (HSPA9) prevents unfolded and dysfunctional protein accumulation, with genetic variants known to be pathogenic. Here, we determined the genetic cause of Even-Plus syndrome (OMIM: 616854) in a Chinese family. METHODS We collected samples from two affected and two normal individuals. Whole-exome sequencing was performed to identify their genetic profiles. Potential variants were validated using Sanger sequencing. Assisted reproduction with mutation-free embryos successfully blocked the transmission of mutations. RESULTS We identified novel inherited pathogenic complex heterozygous variations in the HSPA9 gene in the two affected fetuses. Three-dimensional spatial simulation of the HSPA9 protein after prediction of the mutated RNA splicing pattern abolished part of the substrate-binding domain of the protein. According to ACMG guidelines, c. 1822-1G>A and c. 1411-3T>G were classified as pathogenic and likely pathogenic, respectively. Mutation-free embryos were selected for transplantation and reconfirmed to possess no mutations. A healthy daughter was successfully born into the family. CONCLUSIONS This study is the first to report complex heterozygous variations in the HSPA9 gene that influence alternative splicing in early pregnancy. Our findings expand on the mutational spectrum leading to Even-Plus syndrome and provide a basis for genetic counseling and future embryonic studies.
Collapse
Affiliation(s)
- Xiaoxia Chang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Department of Obstetrics and Gynecology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chunmin Ji
- Department of Obstetrics and Gynecology, Air Force Hospital of Eastern Theater, Nanjing, Jiangsu Province, China
| | - Ting Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Huan Huang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Department of Obstetrics and Gynecology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
3
|
Saetang J, Sukkapat P, Mittal A, Julamanee J, Khopanlert W, Maneechai K, Nazeer RA, Sangkhathat S, Benjakul S. Proteome Analysis of the Antiproliferative Activity of the Novel Chitooligosaccharide-Gallic Acid Conjugate against the SW620 Colon Cancer Cell Line. Biomedicines 2023; 11:1683. [PMID: 37371778 PMCID: PMC10296375 DOI: 10.3390/biomedicines11061683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Chitooligosaccharide (COS) and gallic acid (GA) are natural compounds with anti-cancer properties, and their conjugate (COS-GA) has several biological activities. Herein, the anti-cancer activity of COS-GA in SW620 colon cancer cells was investigated. MTT assay was used to evaluate cell viability after treatment with 62.5, 122, and 250 µg/mL of COS, GA, and COS-GA for 24 and 48 h. The number of apoptotic cells was determined using flow cytometry. Proteomic analysis was used to explore the mechanisms of action of different compounds. COS-GA and GA showed a stronger anti-cancer effect than COS by reducing SW620 cell proliferation at 125 and 250 µg/mL within 24 h. Flow cytometry revealed 20% apoptosis after COS-GA treatment for 24 h. Thus, GA majorly contributed to the enhanced anti-cancer activity of COS via conjugation. Proteomic analysis revealed alterations in protein translation and DNA duplication in the COS group and the structural constituents of the cytoskeleton, intermediate filament organization, the mitochondrial nucleoid, and glycolytic processes in the COS-GA group. Anti-cancer-activity-related proteins were altered, including CLTA, HSPA9, HIST2H2BF, KRT18, HINT1, DSP, and VIM. Overall, the COS-GA conjugate can serve as a potential anti-cancer agent for the safe and effective treatment of colon cancer.
Collapse
Affiliation(s)
- Jirakrit Saetang
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.S.); (P.S.); (A.M.)
| | - Phutthipong Sukkapat
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.S.); (P.S.); (A.M.)
| | - Ajay Mittal
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.S.); (P.S.); (A.M.)
| | - Jakrawadee Julamanee
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.J.); (W.K.); (K.M.)
| | - Wannakorn Khopanlert
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.J.); (W.K.); (K.M.)
| | - Kajornkiat Maneechai
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.J.); (W.K.); (K.M.)
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India;
| | - Surasak Sangkhathat
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand;
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.S.); (P.S.); (A.M.)
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
4
|
Wang J, Huang H, Zhang X, Ma H. LOXL1‑AS1 promotes thymoma and thymic carcinoma progression by regulating miR‑525‑5p‑ HSPA9. Oncol Rep 2021; 45:117. [PMID: 33907842 PMCID: PMC8107651 DOI: 10.3892/or.2021.8068] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/12/2021] [Indexed: 12/14/2022] Open
Abstract
Due to the lack of specific symptoms in early thymic epithelial tumours (TETs), patients are mostly in the advanced stage at the time of presentation. The aim of the present study was to explore the mechanism by which the long noncoding RNA (lncRNA) LOXL1‑AS1 affects thymoma and thymic carcinoma progression by targeting the miR‑525‑5p‑HSPA9 axis. Bioinformatics was used to analyse the process of LOXL1‑AS1 targeting miR‑525‑5p‑HSPA9 and its expression characteristics in TET. The relationships between LOXL1‑AS1, miR‑525‑5p, HSPA9 and prognosis were analysed. The dual luciferase reporter assay was applied to verify targeting. The gene was knocked down or overexpressed by plasmid transfection. Cell counting kit 8 (CCK‑8) assay, flow cytometry and Transwell assay were used to detect cell viability, apoptosis and invasion ability, respectively. Proteins and RNAs were examined by western blot analysis and qPCR, respectively. A tumour‑burdened assay was used to perform in vivo verification. LOXL1‑AS1 and HSPA9 were overexpressed in thymoma and thymic carcinoma; high levels of LOXL1‑AS1 and HSPA9 were associated with poor prognosis, and there was a significant positive correlation between their levels. Downregulation of miR‑525‑5p expression was also associated with poor prognosis of patients. Clinical trials also demonstrated the same trends. miR‑525‑5p inhibited the expression of HSPA9 protein by targeting the 3'‑untranslated region (UTR) of HSPA9 mRNA. LOXL1‑AS1 promoted the expression of HSPA9 as a sponge targeting miR‑525‑5p. Animal experiment results also showed that knockdown of miR‑525‑5p promoted cancer by promoting the expression of HSPA9. In conclusion, LOXL1‑AS1 and HSPA9 are highly expressed in thymoma and thymic carcinoma; miR‑525‑5p is expressed at low levels in thymoma and thymic carcinoma; and downregulation of miR‑525‑5p is associated with poor prognosis. In summary, this study demonstrates that LOXL1‑AS1 acts as a sponge that targets miR‑525‑5p to promote HSPA9 expression, thereby promoting the growth and invasion and inhibiting apoptosis of thymoma and thymic carcinoma cells.
Collapse
Affiliation(s)
- Jin Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai 200080, P.R. China
| | - Haihua Huang
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai 200080, P.R. China
| | - Xiaomiao Zhang
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai 200080, P.R. China
| | - Haitao Ma
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
5
|
Madkour M, Aboelenin MM, Shakweer WME, Alfarraj S, Alharbi SA, Abdel-Fattah SA, Alagawany M. Early life thermal stress modulates hepatic expression of thermotolerance related genes and physiological responses in two rabbit breeds. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1914207] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Mahmoud Madkour
- Animal Production Department, National Research Centre, Dokki, Egypt
| | | | | | - Saleh Alfarraj
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany & Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sayed A. Abdel-Fattah
- Faculty of Agriculture, Poultry Production Department, Ain Shams University, Cairo, Egypt
| | - Mahmoud Alagawany
- Faculty of Agriculture, Poultry Department, Zagazig University, Zagazig, Egypt
| |
Collapse
|
6
|
Jiao D, Zhang J, Chen P, Guo X, Qiao J, Zhu J, Wang L, Lu Z, Liu Z. HN1L promotes migration and invasion of breast cancer by up-regulating the expression of HMGB1. J Cell Mol Med 2021; 25:397-410. [PMID: 33191617 PMCID: PMC7810958 DOI: 10.1111/jcmm.16090] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/01/2020] [Accepted: 10/11/2020] [Indexed: 02/06/2023] Open
Abstract
Recent reports showed that haematological and neurological expressed 1-like (HN1L) gene participated in tumorigenesis and tumour invasion. However, the expression and role of HN1L in breast cancer remain to be investigated. Here, bioinformatics, western blot and immunohistochemistry were used to detect the expression of HN1L in breast cancer. Wound healing, transwell assay, immunofluorescence assay and mass spectrum were used to explore the role and mechanism of HN1L on the migration and invasion of breast cancer, which was confirmed in vivo using a nude mice model. Results showed that HN1L was significantly over-expressed in breast cancer tissues, which was positively correlated with M metastasis of breast cancer patients. Silencing HN1L significantly inhibited the invasion and metastasis of breast cancer cells in vitro and lung metastasis in nude mice metastasis model of breast cancer. Mechanistically, HN1L interacted with HSPA9 and affected the expression of HMGB1, playing a key role in promoting the invasion and metastasis of breast cancer cell. These results suggested that HN1L was an appealing drug target for breast cancer.
Collapse
Affiliation(s)
- Dechuang Jiao
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Jingyang Zhang
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Ping Chen
- College of Basic Medical SciencesCollaborative Innovation Center of Henan Province for Cancer ChemopreventionZhengzhou UniversityZhengzhouChina
| | - Xuhui Guo
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Jianghua Qiao
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Jiujun Zhu
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Lina Wang
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Zhenduo Lu
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Zhenzhen Liu
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| |
Collapse
|
7
|
Malla WA, Arora R, Khan RIN, Mahajan S, Tiwari AK. Apoptin as a Tumor-Specific Therapeutic Agent: Current Perspective on Mechanism of Action and Delivery Systems. Front Cell Dev Biol 2020; 8:524. [PMID: 32671070 PMCID: PMC7330108 DOI: 10.3389/fcell.2020.00524] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer remains one of the leading causes of death worldwide in humans and animals. Conventional treatment regimens often fail to produce the desired outcome due to disturbances in cell physiology that arise during the process of transformation. Additionally, development of treatment regimens with no or minimum side-effects is one of the thrust areas of modern cancer research. Oncolytic viral gene therapy employs certain viral genes which on ectopic expression find and selectively destroy malignant cells, thereby achieving tumor cell death without harming the normal cells in the neighborhood. Apoptin, encoded by Chicken Infectious Anemia Virus' VP3 gene, is a proline-rich protein capable of inducing apoptosis in cancer cells in a selective manner. In normal cells, the filamentous Apoptin becomes aggregated toward the cell margins, but is eventually degraded by proteasomes without harming the cells. In malignant cells, after activation by phosphorylation by a cancer cell-specific kinase whose identity is disputed, Apoptin accumulates in the nucleus, undergoes aggregation to form multimers, and prevents the dividing cancer cells from repairing their DNA lesions, thereby forcing them to undergo apoptosis. In this review, we discuss the present knowledge about the structure of Apoptin protein, elaborate on its mechanism of action, and summarize various strategies that have been used to deliver it as an anticancer drug in various cancer models.
Collapse
Affiliation(s)
- Waseem Akram Malla
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Richa Arora
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Raja Ishaq Nabi Khan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Sonalika Mahajan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Ashok Kumar Tiwari
- Division of Biological Standardisation, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| |
Collapse
|
8
|
Jeong J, Jin S, Choi H, Kwon JT, Kim J, Kim J, Park ZY, Cho C. Characterization of MAGEG2 with testis-specific expression in mice. Asian J Androl 2018; 19:659-665. [PMID: 27852984 PMCID: PMC5676425 DOI: 10.4103/1008-682x.192033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Male germ cell development is a well-defined process occurring in numerous seminiferous tubules of the testis. Uncovering testicular novel genes related to intrinsic regulation of spermatogenesis is essential for the understanding of spermatogenesis. In the present study, we investigated mouse Mageg2, which belongs to a group of melanoma-associated antigens (MAGEs). Mageg2 is transcribed in the testis specifically, and its expression level is increased at the pachytene spermatocyte stage, indicating that Mageg2 is expressed predominantly in germ cells. We generated an antibody against mouse MAGEG2 for further characterization at the protein level. Immunoblot analysis suggested that MAGEG2 has specific testicular expression and the expression primarily occurred in pachytene spermatocytes. Proteomic analyses demonstrated that mouse MAGEG2 binded to testicular germ cell-specific serine/threonine-protein kinase 31 (STK31) and heat shock protein 9 (HSPA9). Direct binding with both interaction partners was confirmed by co-immunoprecipitation. We found that STK31 and HSPA9 bind MAGEG2 directly but not with each other. Interestingly, MAGEG2 reduced the kinase activity of STK31. Our study suggests that mouse MAGEG2 has at least two functions, including chaperone activity related to HSPA9 and regulation of pachytene spermatocyte-specific kinase, STK31. Altogether, our results provide the first information about MAGEG2 at the transcript and protein levels and suggest its potential molecular functions.
Collapse
Affiliation(s)
- Juri Jeong
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea
| | - Sora Jin
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea
| | - Heejin Choi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea
| | - Jun Tae Kwon
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea
| | - Jihye Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea
| | - Jaehwan Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea
| | - Zee Yong Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea
| | - Chunghee Cho
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea
| |
Collapse
|
9
|
Radons J. The human HSP70 family of chaperones: where do we stand? Cell Stress Chaperones 2016; 21:379-404. [PMID: 26865365 PMCID: PMC4837186 DOI: 10.1007/s12192-016-0676-6] [Citation(s) in RCA: 391] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 01/23/2023] Open
Abstract
The 70-kDa heat shock protein (HSP70) family of molecular chaperones represents one of the most ubiquitous classes of chaperones and is highly conserved in all organisms. Members of the HSP70 family control all aspects of cellular proteostasis such as nascent protein chain folding, protein import into organelles, recovering of proteins from aggregation, and assembly of multi-protein complexes. These chaperones augment organismal survival and longevity in the face of proteotoxic stress by enhancing cell viability and facilitating protein damage repair. Extracellular HSP70s have a number of cytoprotective and immunomodulatory functions, the latter either in the context of facilitating the cross-presentation of immunogenic peptides via major histocompatibility complex (MHC) antigens or in the context of acting as "chaperokines" or stimulators of innate immune responses. Studies have linked the expression of HSP70s to several types of carcinoma, with Hsp70 expression being associated with therapeutic resistance, metastasis, and poor clinical outcome. In malignantly transformed cells, HSP70s protect cells from the proteotoxic stress associated with abnormally rapid proliferation, suppress cellular senescence, and confer resistance to stress-induced apoptosis including protection against cytostatic drugs and radiation therapy. All of the cellular activities of HSP70s depend on their adenosine-5'-triphosphate (ATP)-regulated ability to interact with exposed hydrophobic surfaces of proteins. ATP hydrolysis and adenosine diphosphate (ADP)/ATP exchange are key events for substrate binding and Hsp70 release during folding of nascent polypeptides. Several proteins that bind to distinct subdomains of Hsp70 and consequently modulate the activity of the chaperone have been identified as HSP70 co-chaperones. This review focuses on the regulation, function, and relevance of the molecular Hsp70 chaperone machinery to disease and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Jürgen Radons
- Scientific Consulting International, Mühldorfer Str. 64, 84503, Altötting, Germany.
| |
Collapse
|
10
|
Liu J, Huang W, Lin Y, Bian L, He Y. Identification of proteins interacting with protein kinase C-δ in hyperthermia-induced apoptosis and thermotolerance of Tca8113 cells. Mol Med Rep 2015; 12:3821-3828. [PMID: 26017369 DOI: 10.3892/mmr.2015.3861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 04/30/2015] [Indexed: 11/06/2022] Open
Abstract
The purpose of the present study was to investigate the differential proteins that interact with protein kinase C‑δ (PKC‑δ) in hyperthermia‑induced apoptosis as well as thermotolerance in Tca8113 cells, and furthermore, to investigate the mechanisms of these processes in tumor cells. Activation of PKC‑δ was previously indicated to be involved in the heat sensitivity and thermal resistance of tongue squamous carcinoma cells. Tca8113 cell apoptosis was induced by incubation at 43˚C for 80 min and the thermotolerant Tca8113 cells (TT‑Tca8113) were generated through a gradient temperature‑elevating method. The apoptotic rate of the cells was determined by flow cytometry, while cleavage and activation of PKC‑δ were analyzed by western blot analysis. The proteins that interacted with PKC‑δ in the Tca8113 and TT‑Tca8113 cells were identified by co‑immunoprecipitation coupled with mass spectrometry. Co‑immunoprecipitation analysis followed by electrospray ionization mass spectrometric analysis were utilized to identify the pro‑ and anti‑apoptotic proteins that interacted with PKC‑δ. Significant cell apoptosis was observed in Tca8113 cells following hyperthermia, and the apoptotic rate was significantly higher than that in the control group (P<0.05). Marked PKC‑δ cleavage fragmentation was also identified. By contrast, the apoptotic rate of the TT‑Tca8113 cells was not significantly increased following hyperthermia and no PKC‑δ cleavage fragmentation was observed. Among the proteins interacting with PKC‑δ, 39 were found to be involved in the promotion of apoptosis and 16 in the inhibition of apoptosis of Tca8113 cells; these proteins were known to be involved in the regulation of cell proliferation, apoptosis, transcription and intracellular protein transport. The results of the present study provided evidence that PKC‑δ is a crucial factor in the heat sensitivity and thermal resistance of tongue squamous carcinoma cells and elucidated the underlying molecular basis, which may aid in the improvement of hyperthermic cancer treatments.
Collapse
Affiliation(s)
- Jianqi Liu
- Department of Dental Research, The Affiliated Stomatological Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Wenchuan Huang
- Department of Dental Research, The Affiliated Stomatological Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Yunhong Lin
- Department of Dental Research, The Affiliated Stomatological Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Li Bian
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yongwen He
- Department of Dental Research, The Affiliated Stomatological Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| |
Collapse
|
11
|
Morisaki T, Yashiro M, Kakehashi A, Inagaki A, Kinoshita H, Fukuoka T, Kasashima H, Masuda G, Sakurai K, Kubo N, Muguruma K, Ohira M, Wanibuchi H, Hirakawa K. Comparative proteomics analysis of gastric cancer stem cells. PLoS One 2014; 9:e110736. [PMID: 25379943 PMCID: PMC4224387 DOI: 10.1371/journal.pone.0110736] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 09/16/2014] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are responsible for cancer progression, metastasis, and recurrence. To date, the specific markers of CSCs remain undiscovered. The aim of this study was to identify novel biomarkers of gastric CSCs for clinical diagnosis using proteomics technology. CSC-like SP cells, OCUM-12/SP cells, OCUM-2MD3/SP cells, and their parent OCUM-12 cells and OCUM-2MD3 cells were used in this study. Protein lysates from each cell line were analyzed using QSTAR Elite Liquid Chromatography with Tandem Mass Spectrometry, coupled with isobaric tags for relative and absolute quantitation technology. Candidate proteins detected by proteomics technology were validated by immunohistochemical analysis of 300 gastric cancers. Based on the results of LC-MS/MS, eight proteins, including RBBP6, GLG1, VPS13A, DCTPP1, HSPA9, HSPA4, ALDOA, and KRT18, were up-regulated in both OCUM-12/SP cells and OCUM-2MD3/SP cells when compared to their corresponding parent cells. RT-PCR analysis indicated that the expression level of RBBP6, HSPA4, DCTPP1, HSPA9, VPS13A, ALDOA, GLG1, and CK18 was high in OCUM-12/SP and OCUM-2MD3/SP, in compared with the control of parent OCUM-12 and OCUM-2MD3. These proteins were significantly associated with advanced invasion depth, lymph node metastasis, distant metastasis, or advanced clinical stage. RBBP6, DCTPP1, HSPA4, and ALDOA expression in particular were significantly associated with a poor prognosis in the 300 gastric cancer patients. RBBP6 was determined to be an independent prognostic factor. The motility-stimulating ability of OCUM-12/SP cells and OCUM-2MD3/SP cells was inhibited by RBBP6 siRNA. These findings might suggest that the eight proteins, RBBP6, GLG1, VPS13A, DCTPP1, HSPA9, HSPA4, ALDOA, and KRT18, utilizing comparative proteomics analysis, were perceived to be potential CSC markers of gastric cancer. Of the eight candidate proteins, RBBP6 was suggested to be a promising prognostic biomarker and a therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Tamami Morisaki
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
- Oncology Institute of Geriatrics and Medical Science, Osaka City University Graduate School of Medicine, Osaka, Japan
- * E-mail:
| | - Anna Kakehashi
- Department of Pathology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Azusa Inagaki
- Department of Pathology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Haruhito Kinoshita
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tatsunari Fukuoka
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Kasashima
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Go Masuda
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Katsunobu Sakurai
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Naoshi Kubo
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kazuya Muguruma
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaichi Ohira
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hideki Wanibuchi
- Department of Pathology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|