1
|
Jeong HN, Lee TG, Park HJ, Yang Y, Oh SH, Kang SW, Choi YC. Transcriptome analysis of skeletal muscle in dermatomyositis, polymyositis, and dysferlinopathy, using a bioinformatics approach. Front Neurol 2023; 14:1328547. [PMID: 38125829 PMCID: PMC10731051 DOI: 10.3389/fneur.2023.1328547] [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: 10/26/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Background Polymyositis (PM) and dermatomyositis (DM) are two distinct subgroups of idiopathic inflammatory myopathies. Dysferlinopathy, caused by a dysferlin gene mutation, usually presents in late adolescence with muscle weakness, degenerative muscle changes are often accompanied by inflammatory infiltrates, often resulting in a misdiagnosis as polymyositis. Objective To identify differential biological pathways and hub genes related to polymyositis, dermatomyositis and dysferlinopathy using bioinformatics analysis for understanding the pathomechanisms and providing guidance for therapy development. Methods We analyzed intramuscular ribonucleic acid (RNA) sequencing data from seven dermatomyositis, eight polymyositis, eight dysferlinopathy and five control subjects. Differentially expressed genes (DEGs) were identified by using DESeq2. Enrichment analyses were performed to understand the functions and enriched pathways of DEGs. A protein-protein interaction (PPI) network was constructed, and clarified the gene cluster using the molecular complex detection tool (MCODE) analysis to identify hub genes. Results A total of 1,048, 179 and 3,807 DEGs were detected in DM, PM and dysferlinopathy, respectively. Enrichment analyses revealed that upregulated DEGs were involved in type 1 interferon (IFN1) signaling pathway in DM, antigen processing and presentation of peptide antigen in PM, and cellular response to stimuli in dysferlinopathy. The PPI network and MCODE cluster identified 23 genes related to type 1 interferon signaling pathway in DM, 4 genes (PDIA3, HLA-C, B2M, and TAP1) related to MHC class 1 formation and quality control in PM, and 7 genes (HSPA9, RPTOR, MTOR, LAMTOR1, LAMTOR5, ATP6V0D1, and ATP6V0B) related to cellular response to stress in dysferliniopathy. Conclusion Overexpression of genes related to the IFN1 signaling pathway and major histocompatibility complex (MHC) class I formation was identified in DM and PM, respectively. In dysferlinopathy, overexpression of HSPA9 and the mTORC1 signaling pathway genes was detected.
Collapse
Affiliation(s)
- Ha-Neul Jeong
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Taek Gyu Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Hyung Jun Park
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Yang
- Research Institute of Women's Disease, Sookmyumg Women's University, Seoul, Republic of Korea
| | - Seung-Hun Oh
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, Republic of Korea
| | - Seong-Woong Kang
- Department of Rehabilitation Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Chul Choi
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Meidinna HN, Shefrin S, Sari AN, Zhang H, Dhanjal JK, Kaul SC, Sundar D, Wadhwa R. Identification of a new member of Mortaparib class of inhibitors that target mortalin and PARP1. Front Cell Dev Biol 2022; 10:918970. [PMID: 36172283 PMCID: PMC9510692 DOI: 10.3389/fcell.2022.918970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Mortalin, a heat shock family protein enriched in cancer cells, is known to inactivate tumor suppressor protein p53. Abrogation of mortalin-p53 interaction and reactivation of p53 has been shown to trigger growth arrest/apoptosis in cancer cells and hence, suggested to be useful in cancer therapy. In this premise, we earlier screened a chemical library to identify potential disruptors of mortalin-p53 interaction, and reported two novel synthetic small molecules (5-[1-(4-methoxyphenyl) (1,2,3,4-tetraazol-5-yl)]-4-phenylpyrimidine-2-ylamine) and (4-[(1E)-2-(2-phenylindol-3-yl)-1-azavinyl]-1,2,4-triazole) called Mortaparib and MortaparibPlus, respectively. These compounds were shown to possess anticancer activity that was mediated through targeting mortalin and PARP1 proteins, essential for cancer cell survival and proliferation. Here, we report characterization of the third compound, {4-[(4-amino-5-thiophen-2-yl-1,2,4-triazol-3-yl)sulfanylmethyl]-N-(4-methoxyphenyl)-1,3-thiazol-2-amine}, isolated in the same screening. Extensive computational and molecular analyses suggested that the new compound has the capability to interact with mortalin, p53, and PARP1. We provide evidence that this new compound, although required in high concentration as compared to the earlier two compounds (Mortaparib and MortaparibPlus) and hence called MortaparibMild, also downregulates mortalin and PARP1 expression and functions in multiple ways impeding cancer cell proliferation and migration characteristics. MortaparibMild is a novel candidate anticancer compound that warrants further experimental and clinical attention.
Collapse
Affiliation(s)
- Hazna Noor Meidinna
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, Japan
| | - Seyad Shefrin
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, New Delhi, India
| | - Anissa Nofita Sari
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, Japan
| | - Huayue Zhang
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, Japan
| | - Jaspreet Kaur Dhanjal
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Sunil C. Kaul
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, Japan
| | - Durai Sundar
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, New Delhi, India
- *Correspondence: Durai Sundar, ; Renu Wadhwa,
| | - Renu Wadhwa
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, Japan
- *Correspondence: Durai Sundar, ; Renu Wadhwa,
| |
Collapse
|
3
|
Seth P. Insights Into the Role of Mortalin in Alzheimer’s Disease, Parkinson’s Disease, and HIV-1-Associated Neurocognitive Disorders. Front Cell Dev Biol 2022; 10:903031. [PMID: 35859895 PMCID: PMC9292388 DOI: 10.3389/fcell.2022.903031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Mortalin is a chaperone protein that regulates physiological functions of cells. Its multifactorial role allows cells to survive pathological conditions. Pharmacological, chemical, and siRNA-mediated downregulation of mortalin increases oxidative stress, mitochondrial dysfunction leading to unregulated inflammation. In addition to its well-characterized function in controlling oxidative stress, mitochondrial health, and maintaining physiological balance, recent evidence from human brain autopsies and cell culture–based studies suggests a critical role of mortalin in attenuating the damage seen in several neurodegenerative diseases. Overexpression of mortalin provides an important line of defense against accumulated proteins, inflammation, and neuronal loss, a key characteristic feature observed in neurodegeneration. Neurodegenerative diseases are a group of progressive disorders, sharing pathological features in Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and HIV-associated neurocognitive disorder. Aggregation of insoluble amyloid beta-proteins and neurofibrillary tangles in Alzheimer’s disease are among the leading cause of neuropathology in the brain. Parkinson’s disease is characterized by the degeneration of dopamine neurons in substantia nigra pars compacta. A substantial synaptic loss leading to cognitive decline is the hallmark of HIV-associated neurocognitive disorder (HAND). Brain autopsies and cell culture studies showed reduced expression of mortalin in Alzheimer’s, Parkinson’s, and HAND cases and deciphered the important role of mortalin in brain cells. Here, we discuss mortalin and its regulation and describe how neurotoxic conditions alter the expression of mortalin and modulate its functions. In addition, we also review the neuroprotective role of mortalin under neuropathological conditions. This knowledge showcases the importance of mortalin in diverse brain functions and offers new opportunities for the development of therapeutic targets that can modulate the expression of mortalin using chemical compounds.
Collapse
Affiliation(s)
- Pankaj Seth
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Gurgaon, India
| |
Collapse
|
4
|
Vishwanathan V, D’Silva P. Loss of Function of mtHsp70 Chaperone Variants Leads to Mitochondrial Dysfunction in Congenital Sideroblastic Anemia. Front Cell Dev Biol 2022; 10:847045. [PMID: 35252210 PMCID: PMC8888832 DOI: 10.3389/fcell.2022.847045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Congenital Sideroblastic Anemias (CSA) is a group of rare genetic disorders characterized by the abnormal accumulation of iron in erythrocyte precursors. A common hallmark underlying these pathological conditions is mitochondrial dysfunction due to altered protein homeostasis, heme biosynthesis, and oxidative phosphorylation. A clinical study on congenital sideroblastic anemia has identified mutations in mitochondrial Hsp70 (mtHsp70/Mortalin). Mitochondrial Hsp70 plays a critical role in maintaining mitochondrial function by regulating several pathways, including protein import and folding, and iron-sulfur cluster synthesis. Owing to the structural and functional homology between human and yeast mtHsp70, we have utilized the yeast system to delineate the role of mtHsp70 variants in the etiology of CSA’s. Analogous mutations in yeast mtHsp70 exhibited temperature-sensitive growth phenotypes under non-respiratory and respiratory conditions. In vivo analyses indicate a perturbation in mitochondrial mass and functionality accompanied by an alteration in the organelle network and cellular redox levels. Preliminary in vitro biochemical studies of mtHsp70 mutants suggest impaired import function, altered ATPase activity and substrate interaction. Together, our findings suggest the loss of chaperone activity to be a pivotal factor in the pathophysiology of congenital sideroblastic anemia.
Collapse
|
5
|
Albakova Z, Mangasarova Y, Albakov A, Gorenkova L. HSP70 and HSP90 in Cancer: Cytosolic, Endoplasmic Reticulum and Mitochondrial Chaperones of Tumorigenesis. Front Oncol 2022; 12:829520. [PMID: 35127545 PMCID: PMC8814359 DOI: 10.3389/fonc.2022.829520] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022] Open
Abstract
HSP70 and HSP90 are two powerful chaperone machineries involved in survival and proliferation of tumor cells. Residing in various cellular compartments, HSP70 and HSP90 perform specific functions. Concurrently, HSP70 and HSP90 homologs may also translocate from their primary site under various stress conditions. Herein, we address the current literature on the role of HSP70 and HSP90 chaperone networks in cancer. The goal is to provide a comprehensive review on the functions of cytosolic, mitochondrial and endoplasmic reticulum HSP70 and HSP90 homologs in cancer. Given that high expression of HSP70 and HSP90 enhances tumor development and associates with tumor aggressiveness, further understanding of HSP70 and HSP90 chaperone networks may provide clues for the discoveries of novel anti-cancer therapies.
Collapse
Affiliation(s)
- Zarema Albakova
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
- *Correspondence: Zarema Albakova,
| | | | | | | |
Collapse
|
6
|
Pavlikova N, Sramek J, Jelinek M, Halada P, Kovar J. Markers of acute toxicity of DDT exposure in pancreatic beta-cells determined by a proteomic approach. PLoS One 2020; 15:e0229430. [PMID: 33104727 PMCID: PMC7588079 DOI: 10.1371/journal.pone.0229430] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 10/12/2020] [Indexed: 12/18/2022] Open
Abstract
Many compounds have the potential to harm pancreatic beta-cells; organochlorine pollutants belong to those compounds. In this work, we aimed to find markers of acute toxicity of p,p'-DDT exposure among proteins expressed in NES2Y human pancreatic beta-cells employing 2-D electrophoresis. We exposed NES2Y cells to a high concentration (150 μM, LC96 after 72 hours) of p,p'-DDT for 24 and 30 hours and determined proteins with changed expression using 2-D electrophoresis. We have found 22 proteins that changed their expression. They included proteins involved in ER stress (GRP78, and endoplasmin), mitochondrial proteins (GRP75, ECHM, IDH3A, NDUS1, and NDUS3), proteins involved in the maintenance of the cell morphology (EFHD2, TCPA, NDRG1, and ezrin), and some other proteins (HNRPF, HNRH1, K2C8, vimentin, PBDC1, EF2, PCNA, biliverdin reductase, G3BP1, FRIL, and HSP27). The proteins we have identified may serve as indicators of p,p'-DDT toxicity in beta-cells in future studies, including long-term exposure to environmentally relevant concentrations.
Collapse
Affiliation(s)
- Nela Pavlikova
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Sramek
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Michael Jelinek
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petr Halada
- BioCeV–Institute of Microbiology, The Czech Academy of Sciences, Vestec, Czech Republic
| | - Jan Kovar
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
7
|
PCDHGB7 Increases Chemosensitivity to Carboplatin by Inhibiting HSPA9 via Inducing Apoptosis in Breast Cancer. DISEASE MARKERS 2019; 2019:6131548. [PMID: 31379979 PMCID: PMC6652090 DOI: 10.1155/2019/6131548] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/12/2019] [Accepted: 05/23/2019] [Indexed: 12/31/2022]
Abstract
Breast cancer is one of the most serious cancers worldwide, and chemotherapy resistance frequently drives cancer progression. Triple-negative breast cancer (TNBC) has a high recurrence rate and poor prognosis given its resistance to chemotherapy. In our previous study, we found a remarkable abnormal methylation modification of the PCDHGB7 gene in breast cancer. However, the roles of PCDHGB7 in the progression and treatment of breast cancer are unclear. In this study, we examined the effects of PCDHGB7 on the sensitivity of TNBC cells to carboplatin and investigated the underlying mechanism. By knocking down and overexpressing PCDHGB7 in HS578T and BT549 cells, we confirmed that PCDHGB7 increases TNBC cell chemosensitivity to carboplatin. Mechanistically, we found that PCDHGB7 negatively regulates the expression of HSPA9, uplifting its inhibition on P53 translocation and caspase-3 activation. Thus, we demonstrated that PCDHGB7 increases chemosensitivity of TNBC cells to carboplatin by inhibiting HSPA9 via inducing apoptosis. PCDHGB7 and HSPA9 represent potential therapeutic targets for chemosensitivity in breast cancer.
Collapse
|
8
|
Cheng W, Zhang B, Zikeliyar M, Wang J, Jian H, Wu K, Zhang Y, Ding J. Elevated Mortalin correlates with poor outcome in hepatocellular carcinoma. Ann Diagn Pathol 2019; 42:59-63. [PMID: 31310900 DOI: 10.1016/j.anndiagpath.2019.06.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/17/2019] [Accepted: 06/26/2019] [Indexed: 01/19/2023]
Abstract
Although several lines of evidence existed suggesting that Mortalin was linked with survival in malignant tumors; it has been barely described regarding the prognostic involvement of its expression in hepatocellular carcinoma (HCC). Herein, to understand the prognostic meaning of Mortalin expression, Immunohistochemistry was undertaken to observe the immunohistochemical characteristics of Mortalin on HCC tissue microarray consisting of 90 cases of HCC and its paired normal control dots, followed by detailed statistical analysis with the accompanying clinicopathological variables available, including gender, age, tumor size, differentiation, cirrhosis, vascular invasion, clinical stage, T classification and intrahepatic metastases. Meanwhile, Kaplan-Meier survival curve was plotted to analyze the prognostic difference for patients with high and low expression of Mortalin. It was exhibited that Mortalin was over-expressed in HCC tissues relative to paired normal control and elevated Mortalin significantly correlated with vascular invasion, clinical stage and intrahepatic metastasis. Kaplan-Meier survival analysis revealed that Mortalin was remarkably associated with overall survival and disease-free survival. Multivariate Cox regression analysis showed that expression of Mortalin was an independent prognostic factor in HCC. Collectively, the data we provided here support the prognostic prediction value of Mortalin in HCC.
Collapse
Affiliation(s)
- Wei Cheng
- Department of Hematologic and Oncology, the Center Hospital of Karamay City, Karamay 834000, Xinjiang, China
| | - Bin Zhang
- Department of Hematologic and Oncology, the Center Hospital of Karamay City, Karamay 834000, Xinjiang, China
| | - Mulager Zikeliyar
- Department of Hematologic and Oncology, the Center Hospital of Karamay City, Karamay 834000, Xinjiang, China
| | - Junqiao Wang
- Department of Hematologic and Oncology, the Center Hospital of Karamay City, Karamay 834000, Xinjiang, China
| | - Huiling Jian
- Department of Hematologic and Oncology, the Center Hospital of Karamay City, Karamay 834000, Xinjiang, China
| | - Kexiong Wu
- Department of Hematologic and Oncology, the Center Hospital of Karamay City, Karamay 834000, Xinjiang, China
| | - Yizhi Zhang
- Department of Hematologic and Oncology, the Center Hospital of Karamay City, Karamay 834000, Xinjiang, China
| | - Jinhui Ding
- Department of General Surgery, the Center Hospital of Karamay City, Karamay 834000, Xinjiang, China.
| |
Collapse
|
9
|
Tan C, Dong Y, Wang J, Yang X. Vanadyl acetylacetonate attenuates Aβ pathogenesis in APP/PS1 transgenic mice depending on the intervention stage. NEW J CHEM 2019. [DOI: 10.1039/c9nj00820a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
VAC treatment caused different Grp75 responses before and after Aβ plaque formation.
Collapse
Affiliation(s)
- Chang Tan
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| | - Yaqiong Dong
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| | - Jing Wang
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| | - Xiaoda Yang
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| |
Collapse
|
10
|
Chen QQ, Zhang C, Qin MQ, Li J, Wang H, Xu DX, Wang JQ. Inositol-Requiring Enzyme 1 Alpha Endoribonuclease Specific Inhibitor STF-083010 Alleviates Carbon Tetrachloride Induced Liver Injury and Liver Fibrosis in Mice. Front Pharmacol 2018; 9:1344. [PMID: 30538632 PMCID: PMC6277551 DOI: 10.3389/fphar.2018.01344] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/31/2018] [Indexed: 12/12/2022] Open
Abstract
Accumulating data demonstrated that hepatic endoplasmic reticulum (ER) stress was involved in the pathogenesis of liver fibrosis. Long-term chronic hepatocyte death contributed to liver fibrosis initiation and progression. Previous researches reported that ER stress sensor inositol-requiring enzyme 1 alpha (IRE1α) was first activated in the process of liver fibrosis. STF-083010 was an IRE1α RNase specific inhibitor. This study aimed to explore the effects of STF-083010 on carbon tetrachloride (CCl4)-induced liver injury and subsequent liver fibrosis. Mice were intraperitoneally (i.p.) injected with CCl4 (0.15 ml/kg) for 8 weeks. In STF-083010+CCl4 group, mice were injected with STF-083010 (30 mg/kg, i.p.), twice a week, beginning from the 6th week after CCl4 injection. CCl4 treatment markedly enhanced the levels of serum ALT, TBIL, DBIL and TBA, and STF-083010 had obviously extenuated CCl4-induced exaltation of ALT, DBIL, and TBA levels. CCl4-induced hepatic hydroxyproline and collagen I, major indicators of liver fibrosis, were alleviated by STF-083010. Additionally, CCl4-induced α-smooth muscle actin, a marker for hepatic stellate cells activation, was obviously attenuated in STF-083010-treated mice. Moreover, CCl4-induced upregulation of inflammatory cytokines was suppressed by STF-083010. Mechanistic exploration found that hepatic miR-122 was downregulated in CCl4-treated mice. Hepatic MCP1, CTGF, P4HA1, Col1α1, and Mmp9, target genes of miR-122, were upregulated in CCl4-treated mice. Interestingly, STF-083010 reversed CCl4-induced hepatic miR-122 downregulation. Correspondingly, STF-083010 inhibited CCl4-induced upregulation of miR-122 target genes. This study provides partial evidence that STF-083010 alleviated CCl4-induced liver injury and thus protected against liver fibrosis associated with hepatic miR-122.
Collapse
Affiliation(s)
- Qian-Qian Chen
- The Fourth Affiliated Hospital, Anhui Medical University, Hefei, China.,The Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Cheng Zhang
- Department of Toxicology, Anhui Medical University, Hefei, China
| | - Ming-Qiang Qin
- The Fourth Affiliated Hospital, Anhui Medical University, Hefei, China.,The Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Jian Li
- Department of Toxicology, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Toxicology, Anhui Medical University, Hefei, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, China
| | - Jian-Qing Wang
- The Fourth Affiliated Hospital, Anhui Medical University, Hefei, China.,The Second Affiliated Hospital, Anhui Medical University, Hefei, China
| |
Collapse
|
11
|
Lv Y, Li Y, Zhang D, Zhang A, Guo W, Zhu S. HMGB1-induced asthmatic airway inflammation through GRP75-mediated enhancement of ER-mitochondrial Ca 2+ transfer and ROS increased. J Cell Biochem 2018; 119:4205-4215. [PMID: 29292841 DOI: 10.1002/jcb.26653] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/20/2017] [Indexed: 02/01/2023]
Abstract
Imbalanced T-helper (TH)1/Th2 response contributes significantly to asthma pathogenesis. Our study indicated that HMGB1 play an important role in the release of Th2-associated cytokines of asthma. However, the specific mechanism about HMGB1-induced imbalanced TH1/Th2 response is not known. In vivo, an OVA-induced asthma mouse model was set up and mice treated with anti-HMGB1 IgG. The mice treated with the anti-HMGB1 IgG ameliorated airway hyper-reactivity, disruption of Th1/Th2 balance and the upregulation of GRP75 induced by OVA. In vitro, the exposure of normal human bronchial epithelial cells to HMGB1 resulted in the upregulation of GRP75, proinflammatory cytokine production, enhanced ER-Mitochondrial Ca2+ transfer, and enhancement of reactive oxygen species (ROS). While HMGB1-induced these changes were attenuated by GRP75 siRNA treatment. Sequentially, pretreatment with 2-APB, SKF960365 (SKF) and Ru360 which inhibit ER-Mitochondrial Ca2+ transfer significantly lowered HMGB1-induced the generation of ROS and the release of Th2 cytokines in 16HBE cells. Meanwhile, N-acetylcysteine (NAC) significantly attenuated the HMGB1-mediated pro-inflammatory cytokines release. Therefore, these results indicate that GRP75-mediated ER-Mitochondrial Ca2+ transfer may be an important contributor in imbalanced of Th1/Th2 balance of asthma. Moreover, HMGB1 specifically induces the release of Th2 cytokines through GRP75-mediated enhancement of ER-Mitochondrial Ca2+ transfer and ROS increased.
Collapse
Affiliation(s)
- Yanhua Lv
- Department of Respiratory, Zhongshan city people's hospital, Zhongshan, Guangdong, China
| | - Yanli Li
- Department of Respiratory, Inner Mongolia people's hospital, Hohhot, Inner Mongolia, China
| | - Dandan Zhang
- Department of Respiratory, Zhongshan city people's hospital, Zhongshan, Guangdong, China
| | - Anbing Zhang
- Department of Respiratory, Zhongshan city people's hospital, Zhongshan, Guangdong, China
| | - Weihong Guo
- Department of Respiratory, Zhongshan city people's hospital, Zhongshan, Guangdong, China
| | - Shunfang Zhu
- Department of Respiratory and Critical Care Medicine, Chronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, Gongdong, China
| |
Collapse
|
12
|
Noormohammadi A, Calculli G, Gutierrez-Garcia R, Khodakarami A, Koyuncu S, Vilchez D. Mechanisms of protein homeostasis (proteostasis) maintain stem cell identity in mammalian pluripotent stem cells. Cell Mol Life Sci 2018; 75:275-290. [PMID: 28748323 PMCID: PMC11105389 DOI: 10.1007/s00018-017-2602-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/13/2017] [Accepted: 07/24/2017] [Indexed: 01/10/2023]
Abstract
Protein homeostasis, or proteostasis, is essential for cell function, development, and organismal viability. The composition of the proteome is adjusted to the specific requirements of a particular cell type and status. Moreover, multiple metabolic and environmental conditions challenge the integrity of the proteome. To maintain the quality of the proteome, the proteostasis network monitors proteins from their synthesis through their degradation. Whereas somatic stem cells lose their ability to maintain proteostasis with age, immortal pluripotent stem cells exhibit a stringent proteostasis network associated with their biological function and intrinsic characteristics. Moreover, growing evidence indicates that enhanced proteostasis mechanisms play a central role in immortality and cell fate decisions of pluripotent stem cells. Here, we will review new insights into the melding fields of proteostasis and pluripotency and their implications for the understanding of organismal development and survival.
Collapse
Affiliation(s)
- Alireza Noormohammadi
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - Giuseppe Calculli
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - Ricardo Gutierrez-Garcia
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - Amirabbas Khodakarami
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - Seda Koyuncu
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - David Vilchez
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany.
| |
Collapse
|
13
|
Mortalin deficiency suppresses fibrosis and induces apoptosis in keloid spheroids. Sci Rep 2017; 7:12957. [PMID: 29021584 PMCID: PMC5636810 DOI: 10.1038/s41598-017-13485-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 09/26/2017] [Indexed: 12/02/2022] Open
Abstract
Mortalin (Mot) is a mitochondrial chaperone of the heat shock protein 70 family and it’s pro-proliferative and anti-apoptosis functions could be associated with keloid pathogenesis, and blocking of mortalin and its interaction with p53 might be a potential novel target for the treatment of keloid. Therefore, we generated mortalin-specific small hairpin (sh) RNAs (dE1-RGD/GFP/shMot) and introduced into keloid spheroids for examination of its apoptotic and anti-fibrotic effect. On keloid tissues, mortalin expression was higher than adjacent normal tissues and it’s protein expressions were activated keloid fibroblasts (KFs). After primary keloid spheroid were transduced with dE1-RGD/GFP/shMot for knockdown of mortalin, expression of type I, III collagen, fibronectin, and elastin was significantly reduced and transforming growth factor-β1, epidermal growth factor receptor (EGFR), Extracellular Signal-Regulated Kinases 1 and 2 (Erk 1/2), and Smad 2/3 complex protein expression were decreased. In addition, increased TUNEL activities and cytochrome C were observed. Further, for examine of mortalin and p53 interaction, we performed immunofluorescence analysis. Knockdown of mortalin relocated p53 to the cell nucleus in primary keloid spheroids by dE1-RGD/GFP/shMot transduction. These results support the utility of knockdown of mortalin to induce apoptosis and reduce ECMs expression in keloid spheroid, which may be highly beneficial in treating keloids.
Collapse
|
14
|
Huang MB, Gonzalez RR, Lillard J, Bond VC. Secretion modification region-derived peptide blocks exosome release and mediates cell cycle arrest in breast cancer cells. Oncotarget 2017; 8:11302-11315. [PMID: 28076321 PMCID: PMC5355266 DOI: 10.18632/oncotarget.14513] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 12/24/2016] [Indexed: 01/27/2023] Open
Abstract
PURPOSE Discovery and development of a novel anticancer PEG-SMR-Clu peptide to prevent breast cancer metastasis. How breast cancer cells and primary mammary epithelial cells interact and communicate with each other to promote tumorigenesis and how to prevent tumor metastasis has long been a concern of researchers. Cancer cells secrete exosomes containing proteins and RNA. These factors can influence tumor development by directly targeting cancer cells and tumor stroma. In this study, we determined the effects of a peptide as an inhibitor of exosome secretion on breast tumors. We developed a peptide derived from the Secretion Modification Region (SMR) of HIV-1 Nef protein that was modified with PEG on the N-terminus and with a Clusterin (Clu)-binding peptide on the C-terminus. Attachment of PEG to the SMR peptide, termed PEGylation, offers improved water solubility and stability as well as reduced clearance through the kidneys, leading to a longer circulation time. The 12-mer Clu-binding peptide plays multiple roles in tumor development and metastasis. The Clu peptide can be detected by antibody in vivo, thus it has the potential to be used to monitor tumor status and treatment efficacy in animal studies and eventually in cancer patients. RESULTS PEG-SMRwt-Clu and PEG-SMRwt peptides inhibited the growth of both of MCF-7 (estrogen responsive, ER+) and MDA-MD-231 (estrogen non-responsive, ER-) human breast cancer cells in a dose and time-dependent manner, without inducing cytotoxic effects. The SMRwt peptide, combined with paclitaxel, induced G2/M phase cell cycle arrest on MCF-7 and MDA-MB-231 cells but did not promote apoptosis. PEG-SMRwt-Clu peptide treatment blocked exosome release from both MCF-7 and MDA-MB-231 cells. This effect was blocked by knockdown of the chaperone protein mortalin by either antibody or siRNA. MATERIALS AND METHODS MCF-7 and MDA-MB-231 breast tumor cells were treated with PEG-SMR-Clu peptide alone and in combination with paclitaxel and cisplatin. Cell proliferation and viabilty were determined via cell cycle analysis using Cellometer imaging cytometry, Annexin V and MTT assays. The effects of the PEG-SMR-Clu peptide on tumor exosome release were determined by testing isolated exosome fractions, for (i) expression of CD63 and Alix proteins by Western blotting, (ii) NanoSight nanoparticle tracking analysis (NTA 10) to measure exosomes size and concentration, and (iii) measurement of acetylcholinesterase (AchE) for exosome specific enzyme activity. CONCLUSIONS PEG-SMRwt-CLU peptides inhibited the growth of human breast cancer cells and blocked tumor exosome release in vitro. The peptide alone did not cause increased cytotoxicity or apoptosis induction, but did cause cell cycle G2/M phase arrest in both estrogen responsive and non-responsive breast cancer cells. These data suggest a potential therapeutic value of SMR to prevent breast cancer metastasis and as an adjuvant for the chemotherapeutic treatment of human breast cancer.
Collapse
Affiliation(s)
- Ming-Bo Huang
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, 30310, USA
| | - Ruben R Gonzalez
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, 30310, USA
| | - James Lillard
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, 30310, USA
| | - Vincent C Bond
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, 30310, USA
| |
Collapse
|
15
|
Salvianolic acid B inhibits mitochondrial dysfunction by up-regulating mortalin. Sci Rep 2017; 7:43097. [PMID: 28251987 PMCID: PMC5333085 DOI: 10.1038/srep43097] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 01/19/2017] [Indexed: 12/11/2022] Open
Abstract
Salvianolic acid B is an antioxidative ingredient derived from Radix Salviae miltiorrhizae that has been widely used to treat liver diseases. However, the therapeutic mechanism underlying Salvianolic acid B has remained largely unknown. Our studies verified that Salvianolic acid B efficiently blocked mitochondrial deformation and dysfunction induced by H2O2 in the human hepatocyte cell line HL7702. Mortalin, a mitochondrial molecular chaperone, maintains mitochondrial morphology stabilization and function integrity. Previous results showed that mortalin overexpression has been observed in hematoma carcinoma cells and that mortalin maintains mitochondrial homeostasis and antagonizes oxidative stress damage. We found that Salvianolic acid B significantly up-regulated mortalin protein expression levels. In addition, Salvianolic acid B lost the function of preventing mitochondrial deformation and dysfunction induced by oxidative stress under mortalin knockdown conditions. We further found that mortalin overexpression increases the mRNA expression of mitofusin-related factor Mfn1 and mitofission-related factor hFis1. In conclusion, Salvianolic acid B maintains the mitochondrial structure stabilization and functional integrity by up-regulating mortalin, which may be associated with increased mitofusin factor Mfn1 and reduced mitofission factor hFis1.
Collapse
|
16
|
Liu T, Krysiak K, Shirai CL, Kim S, Shao J, Ndonwi M, Walter MJ. Knockdown of HSPA9 induces TP53-dependent apoptosis in human hematopoietic progenitor cells. PLoS One 2017; 12:e0170470. [PMID: 28178280 PMCID: PMC5298293 DOI: 10.1371/journal.pone.0170470] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 01/05/2017] [Indexed: 11/19/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are the most common adult myeloid blood cancers in the US. Patients have increased apoptosis in their bone marrow cells leading to low peripheral blood counts. The full complement of gene mutations that contribute to increased apoptosis in MDS remains unknown. Up to 25% of MDS patients harbor and acquired interstitial deletion on the long arm of chromosome 5 [del(5q)], creating haploinsufficiency for a large set of genes including HSPA9. Knockdown of HSPA9 in primary human CD34+ hematopoietic progenitor cells significantly inhibits growth and increases apoptosis. We show here that HSPA9 knockdown is associated with increased TP53 expression and activity, resulting in increased expression of target genes BAX and p21. HSPA9 protein interacts with TP53 in CD34+ cells and knockdown of HSPA9 increases nuclear TP53 levels, providing a possible mechanism for regulation of TP53 by HSPA9 haploinsufficiency in hematopoietic cells. Concurrent knockdown of TP53 and HSPA9 rescued the increased apoptosis observed in CD34+ cells following knockdown of HSPA9. Reduction of HSPA9 below 50% results in severe inhibition of cell growth, suggesting that del(5q) cells may be preferentially sensitive to further reductions of HSPA9 below 50%, thus providing a genetic vulnerability to del(5q) cells. Treatment of bone marrow cells with MKT-077, an HSPA9 inhibitor, induced apoptosis in a higher percentage of cells from MDS patients with del(5q) compared to non-del(5q) MDS patients and normal donor cells. Collectively, these findings indicate that reduced levels of HSPA9 may contribute to TP53 activation and increased apoptosis observed in del(5q)-associated MDS.
Collapse
Affiliation(s)
- Tuoen Liu
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kilannin Krysiak
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Cara Lunn Shirai
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sanghyun Kim
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jin Shao
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Matthew Ndonwi
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Matthew J Walter
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
17
|
Tubbs E, Rieusset J. Study of Endoplasmic Reticulum and Mitochondria Interactions by In Situ Proximity Ligation Assay in Fixed Cells. J Vis Exp 2016. [PMID: 28060261 DOI: 10.3791/54899] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Structural interactions between the endoplasmic reticular (ER) and mitochondrial membranes, in domains known as mitochondria-associated membranes (MAM), are crucial hubs for cellular signaling and cell fate. Particularly, these inter-organelle contact sites allow the transfer of calcium from the ER to mitochondria through the voltage-dependent anion channel (VDAC)/glucose-regulated protein 75 (GRP75)/inositol 1,4,5-triphosphate receptor (IP3R) calcium channeling complex. While this subcellular compartment is under intense investigation in both physiological and pathological conditions, no simple and sensitive method exists to quantify the endogenous amount of ER-mitochondria contact in cells. Similarly, MAMs are highly dynamic structures, and there is no suitable approach to follow modifications of ER-mitochondria interactions without protein overexpression. Here, we report an optimized protocol based on the use of an in situ proximity ligation assay to visualize and quantify endogenous ER-mitochondria interactions in fixed cells by using the close proximity between proteins of the outer mitochondrial membrane (VDAC1) and of the ER membrane (IP3R1) at the MAM interface. Similar in situ proximity ligation experiments can also be performed with the GRP75/IP3R1 and cyclophilin D/IP3R1 pairs of antibodies. This assay provides several advantages over other imaging procedures, as it is highly specific, sensitive, and suitable to multiple-condition testing. Therefore, the use of this in situ proximity ligation assay should be helpful to better understand the physiological regulations of ER-mitochondria interactions, as well as their role in pathological contexts.
Collapse
Affiliation(s)
- Emily Tubbs
- Lund University Diabetes Centre, Lund University;
| | - Jennifer Rieusset
- INSERM UMR-1060, CarMeN Laboratory, Lyon 1 University, INRA U1235, INSA of Lyon, Rockefeller and Charles Merieux Lyon-Sud Medical Universities
| |
Collapse
|
18
|
Control of mitochondrial function and cell growth by the atypical cadherin Fat1. Nature 2016; 539:575-578. [PMID: 27828948 DOI: 10.1038/nature20170] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 10/19/2016] [Indexed: 12/20/2022]
Abstract
Mitochondrial products such as ATP, reactive oxygen species, and aspartate are key regulators of cellular metabolism and growth. Abnormal mitochondrial function compromises integrated growth-related processes such as development and tissue repair, as well as homeostatic mechanisms that counteract ageing and neurodegeneration, cardiovascular disease, and cancer. Physiologic mechanisms that control mitochondrial activity in such settings remain incompletely understood. Here we show that the atypical Fat1 cadherin acts as a molecular 'brake' on mitochondrial respiration that regulates vascular smooth muscle cell (SMC) proliferation after arterial injury. Fragments of Fat1 accumulate in SMC mitochondria, and the Fat1 intracellular domain interacts with multiple mitochondrial proteins, including critical factors associated with the inner mitochondrial membrane. SMCs lacking Fat1 (Fat1KO) grow faster, consume more oxygen for ATP production, and contain more aspartate. Notably, expression in Fat1KO cells of a modified Fat1 intracellular domain that localizes exclusively to mitochondria largely normalizes oxygen consumption, and the growth advantage of these cells can be suppressed by inhibition of mitochondrial respiration, which suggest that a Fat1-mediated growth control mechanism is intrinsic to mitochondria. Consistent with this idea, Fat1 species associate with multiple respiratory complexes, and Fat1 deletion both increases the activity of complexes I and II and promotes the formation of complex-I-containing supercomplexes. In vivo, Fat1 is expressed in injured human and mouse arteries, and inactivation of SMC Fat1 in mice potentiates the response to vascular damage, with markedly increased medial hyperplasia and neointimal growth, and evidence of higher SMC mitochondrial respiration. These studies suggest that Fat1 controls mitochondrial activity to restrain cell growth during the reparative, proliferative state induced by vascular injury. Given recent reports linking Fat1 to cancer, abnormal kidney and muscle development, and neuropsychiatric disease, this Fat1 function may have importance in other settings of altered cell growth and metabolism.
Collapse
|
19
|
Liu Q, Huang X, Zhao D, Han K, Liu Y, Yang J, Bi K, Li Y. Identification of heat shock protein A9 as a Tembusu virus binding protein on DF-1 cells. Virus Res 2016; 227:110-114. [PMID: 27693918 DOI: 10.1016/j.virusres.2016.09.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 09/25/2016] [Accepted: 09/28/2016] [Indexed: 10/20/2022]
Abstract
This study attempts to identify receptor elements for Tembusu virus (TMUV) on DF-1 cells. Using co-immunoprecipitation and virus overlay protein binding assays, we identified a TMUV-binding protein of approximately 70-kDa on DF-1 cell membranes. Mass spectroscopy identified the protein to be heat shock protein (HSP) A9, which was reconfirmed by an anti-HSPA9 antibody. Indirect immunofluorescence demonstrated a significant degree of colocalization between HSPA9 and TMUV on cell surface. Additionally, an antibody against HSPA9 could inhibit TMUV infection in DF-1 cells in a dose-dependent manner. These results might suggest that HSPA9 is a putative receptor for TMUV.
Collapse
Affiliation(s)
- Qingtao Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, Jiangsu, China
| | - Xinmei Huang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, Jiangsu, China
| | - Dongmin Zhao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, Jiangsu, China
| | - Kaikai Han
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, Jiangsu, China
| | - Yuzhuo Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, Jiangsu, China
| | - Jing Yang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, Jiangsu, China
| | - Keran Bi
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, Jiangsu, China
| | - Yin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, Jiangsu, China.
| |
Collapse
|
20
|
Transcriptional Response to Acute Thermal Exposure in Juvenile Chinook Salmon Determined by RNAseq. G3-GENES GENOMES GENETICS 2015; 5:1335-49. [PMID: 25911227 PMCID: PMC4502368 DOI: 10.1534/g3.115.017699] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Thermal exposure is a serious and growing challenge facing fish species worldwide. Chinook salmon (Oncorhynchus tshawytscha) living in the southern portion of their native range are particularly likely to encounter warmer water due to a confluence of factors. River alterations have increased the likelihood that juveniles will be exposed to warm water temperatures during their freshwater life stage, which can negatively impact survival, growth, and development and pose a threat to dwindling salmon populations. To better understand how acute thermal exposure affects the biology of salmon, we performed a transcriptional analysis of gill tissue from Chinook salmon juveniles reared at 12° and exposed acutely to water temperatures ranging from ideal to potentially lethal (12° to 25°). Reverse-transcribed RNA libraries were sequenced on the Illumina HiSeq2000 platform and a de novo reference transcriptome was created. Differentially expressed transcripts were annotated using Blast2GO and relevant gene clusters were identified. In addition to a high degree of downregulation of a wide range of genes, we found upregulation of genes involved in protein folding/rescue, protein degradation, cell death, oxidative stress, metabolism, inflammation/immunity, transcription/translation, ion transport, cell cycle/growth, cell signaling, cellular trafficking, and structure/cytoskeleton. These results demonstrate the complex multi-modal cellular response to thermal stress in juvenile salmon.
Collapse
|
21
|
Abstract
SIGNIFICANCE Stem cells are characterized by the properties of self-renewal and the ability to differentiate into multiple cell types, and thus maintain tissue homeostasis. Reactive oxygen species (ROS) are a natural byproduct of aerobic metabolism and have roles in cell signaling. Regulation of ROS has a vital role in maintaining "stemness" and differentiation of the stem cells, as well as in progression of stem-cell-associated diseases. RECENT ADVANCES As of late, much research has been done on the adverse effects of ROS in stem cells. However, recently it has become apparent that in some cases redox status of the stem cell does have a role in maintaining its identity as such. Both pluripotent and multipotent stem cell types have been reported to possess enzymatic and nonenzymatic mechanisms for detoxification of ROS and to correct oxidative damage to the genome as well as the proteome. CRITICAL ISSUES Although context dependent and somewhat varied among different stem cell types, the correlation seems to exist between antioxidant defense level and stem cell fate change (i.e., proliferation, differentiation, and death). Changes in stem cell redox regulation may affect the pathogenesis of various human diseases. FUTURE DIRECTIONS Dissecting the defined roles of ROS in distinct stem cell types will greatly enhance their basic and translational applications. Here, we discuss the various roles of ROS in adult, embryonic, and induced pluripotent stem cells.
Collapse
Affiliation(s)
- Pooja Chaudhari
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | | | | |
Collapse
|
22
|
Bhagwat SR, Redij T, Phalnikar K, Nayak S, Iyer S, Gadkar S, Chaudhari U, Kholkute SD, Sachdeva G. Cell surfactomes of two endometrial epithelial cell lines that differ in their adhesiveness to embryonic cells. Mol Reprod Dev 2014; 81:326-40. [DOI: 10.1002/mrd.22301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/06/2014] [Indexed: 12/16/2022]
Affiliation(s)
- Sonali R. Bhagwat
- Primate Biology Laboratory; National Institute for Research in Reproductive Health, Indian Council of Medical Research; Mumbai India
| | - Tejashree Redij
- Primate Biology Laboratory; National Institute for Research in Reproductive Health, Indian Council of Medical Research; Mumbai India
| | - Kruttika Phalnikar
- Primate Biology Laboratory; National Institute for Research in Reproductive Health, Indian Council of Medical Research; Mumbai India
| | - Sumeet Nayak
- Primate Biology Laboratory; National Institute for Research in Reproductive Health, Indian Council of Medical Research; Mumbai India
| | - Swati Iyer
- Primate Biology Laboratory; National Institute for Research in Reproductive Health, Indian Council of Medical Research; Mumbai India
| | - Sushama Gadkar
- Primate Biology Laboratory; National Institute for Research in Reproductive Health, Indian Council of Medical Research; Mumbai India
| | - Uddhav Chaudhari
- Primate Biology Laboratory; National Institute for Research in Reproductive Health, Indian Council of Medical Research; Mumbai India
| | - Sanjeeva D. Kholkute
- Primate Biology Laboratory; National Institute for Research in Reproductive Health, Indian Council of Medical Research; Mumbai India
| | - Geetanjali Sachdeva
- Primate Biology Laboratory; National Institute for Research in Reproductive Health, Indian Council of Medical Research; Mumbai India
| |
Collapse
|
23
|
Peng C, Yang P, Cui Y, He M, Liang L, Di Y. HSPA9 overexpression inhibits apoptin-induced apoptosis in the HepG2 cell line. Oncol Rep 2013; 29:2431-7. [PMID: 23589050 DOI: 10.3892/or.2013.2399] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 03/15/2013] [Indexed: 11/06/2022] Open
Abstract
Apoptin, a small protein derived from chicken anemia virus, possesses the capacity to specifically kill tumor cells while leaving normal cells intact. Previous studies have indicated that the subcellular localization of apoptin appears to be crucial for this tumor-selective activity. Apoptin resides in the cytoplasm of normal cells; however, in cancer cells it translocates into the nucleus. In the present study, purified prokaryotic native His-apoptin served as a bait for capturing apoptin-associated proteins in both a hepatoma carcinoma cell line (HepG2) and a human fetal liver cell line (L-02). The captured proteins obtained from a pull-down assay were separated by two-dimensional gel electrophoresis. Mass spectrometry was employed to detect the effect of HSPA9 overexpression (one of the interacting proteins with apoptin in vitro) and downregulation of HSPA9 on HepG2 cells. The data revealed that HSPA9 overexpression resulted in partial distribution of apoptin in the cytoplasm. Notably, HSPA9 overexpression markedly decreased the apoptosis rate of HepG2 cells from 41.2 to 31.7%, while the downregulation of HSPA9 using small interfering RNA significantly enhanced the apoptosis of HepG2 cells. Our results suggest new insights into the localization mechanism of apoptin which is tightly associated with HSPA9 overexpression and its crucial role in cellular apoptosis both in a tumor cell line (HepG2) and a normal cell line (L-02). These findings shed new light on the elucidation of the underlying mechanism of anticancer action of apoptin.
Collapse
Affiliation(s)
- Chuanmei Peng
- Clinical Laboratory of Yanan Hospital of Kunming, and Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming 650051, PR China
| | | | | | | | | | | |
Collapse
|
24
|
Li B, Reed JC, Kim HR, Chae HJ. Proteomic profiling of differentially expressed proteins from Bax inhibitor-1 knockout and wild type mice. Mol Cells 2012; 34:15-23. [PMID: 22736268 PMCID: PMC3887783 DOI: 10.1007/s10059-012-0001-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Revised: 04/04/2012] [Accepted: 05/12/2012] [Indexed: 02/02/2023] Open
Abstract
Bax inhibitor-1 (BI-1) is an anti-apoptotic protein located in the endoplasmic reticulum (ER). The role of BI-1 has been studied in different physiopathological models including ischemia, diabetes, liver regeneration and cancer. However, fundamental knowledge about the effects of BI-1 deletion on the proteome is lacking. To further explore this protein, we compared the levels of different proteins in bi-1 (-/-) and bi-1 (+/+) mouse tissues by two-dimensional electrophoresis (2-DE) and mass spectrometry (MS). In several bi-1 (-/-) mice, glucose-regulated protein 75 (GRP75/mortalin/ PBP74/mthsp70), peroxiredoxin6 (Prx6) and fumarylacetoacetate hydrolase (FAH) showed a pI shift that could be attributed to post-translational modifications. Selenium-binding protein 2 (SBP2) and ferritin light chain 1 levels were significantly increased. Phosphatidylethanolamine-binding protein-1 (PEBP-1) was dramatically decreased in bi-1 (-/-) mice, which was confirmed by Western blotting. The phosphorylation of GRP75, Prx6 and FAH were compared between bi-1 (+/+) and bi-1 (-/-) mice using liver tissue lysates. Of these three proteins, only one exhibited modified phosphorylation; Tyr phosphorylation of Prx6 was increased in bi-1 (-/-) mice. Our protein profiling results provide fundamental knowledge about the physiopathological function of BI-1.
Collapse
Affiliation(s)
- Bo Li
- Department of Pharmacology and Cardiovascular Research Center, Chonbuk National University, Jeonju 561-182,
Korea
| | - John C. Reed
- Burnham Institute for Medical Research, California 92037,
USA
| | - Hyung-Ryong Kim
- Department of Dental Pharmacology, School of Dentistry, Wonkwang University, Iksan 570-749,
Korea
| | - Han-Jung Chae
- Department of Pharmacology and Cardiovascular Research Center, Chonbuk National University, Jeonju 561-182,
Korea
- Research Center for Pulmonary Disorders, Chonbuk National University Hospital, Jeonju 561-182,
Korea
| |
Collapse
|
25
|
Gestl EE, Anne Böttger S. Cytoplasmic sequestration of the tumor suppressor p53 by a heat shock protein 70 family member, mortalin, in human colorectal adenocarcinoma cell lines. Biochem Biophys Res Commun 2012; 423:411-6. [DOI: 10.1016/j.bbrc.2012.05.139] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 05/26/2012] [Indexed: 02/04/2023]
|
26
|
Londono C, Osorio C, Gama V, Alzate O. Mortalin, apoptosis, and neurodegeneration. Biomolecules 2012; 2:143-64. [PMID: 24970131 PMCID: PMC4030873 DOI: 10.3390/biom2010143] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 02/22/2012] [Accepted: 02/23/2012] [Indexed: 02/01/2023] Open
Abstract
Mortalin is a highly conserved heat-shock chaperone usually found in multiple subcellular locations. It has several binding partners and has been implicated in various functions ranging from stress response, control of cell proliferation, and inhibition/prevention of apoptosis. The activity of this protein involves different structural and functional mechanisms, and minor alterations in its expression level may lead to serious biological consequences, including neurodegeneration. In this article we review the most current data associated with mortalin's binding partners and how these protein-protein interactions may be implicated in apoptosis and neurodegeneration. A complete understanding of the molecular pathways in which mortalin is involved is important for the development of therapeutic strategies for cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Carolina Londono
- Systems Proteomics Center Laboratory, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, Escuela de Medicina, Universidad Pontificia Bolivariana, Medellín, Colombia.
| | - Cristina Osorio
- Systems Proteomics Center Laboratory and Program in Molecular Biology and Biotechnology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Vivian Gama
- Neuroscience Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Oscar Alzate
- Systems Proteomics Center Laboratory, Department of Cell and Developmental Biology, Program in Molecular Biology and Biotechnology and Department of Neurology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, Escuela de Medicina, Universidad Pontificia Bolivariana, Medellin, Colombia.
| |
Collapse
|
27
|
Secretion modification region-derived peptide disrupts HIV-1 Nef's interaction with mortalin and blocks virus and Nef exosome release. J Virol 2011; 86:406-19. [PMID: 22013042 DOI: 10.1128/jvi.05720-11] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nef is secreted from infected cells in exosomes and is found in abundance in the sera of HIV-infected individuals. Secreted exosomal Nef (exNef) induces apoptosis in uninfected CD4⁺ T cells and may be a key component of HIV pathogenesis. The exosomal pathway has been implicated in HIV-1 virus release, suggesting a possible link between these two viral processes. However, the underlying mechanisms and cellular components of exNef secretion have not been elucidated. We have previously described a Nef motif, the secretion modification region (SMR; amino acids 66 to 70), that is required for exNef secretion. In silico modeling data suggest that this motif can form a putative binding pocket. We hypothesized that the Nef SMR binds a cellular protein involved in protein trafficking and that inhibition of this interaction would abrogate exNef secretion. By using tandem mass spectrometry and coimmunoprecipitation with a novel SMR-based peptide (SMRwt) that blocks exNef secretion and HIV-1 virus release, we identified mortalin as an SMR-specific cellular protein. A second set of coimmunoprecipitation experiments with full-length Nef confirmed that mortalin interacts with Nef via Nef's SMR motif and that this interaction is disrupted by the SMRwt peptide. Overexpression and microRNA knockdown of mortalin revealed a positive correlation between exNef secretion levels and mortalin protein expression. Using antibody inhibition we demonstrated that the Nef/mortalin interaction is necessary for exNef secretion. Taken together, this work constitutes a significant step in understanding the underlying mechanism of exNef secretion, identifies a novel host-pathogen interaction, and introduces an HIV-derived peptide with antiviral properties.
Collapse
|
28
|
Nuclear GRP75 binds retinoic acid receptors to promote neuronal differentiation of neuroblastoma. PLoS One 2011; 6:e26236. [PMID: 22022577 PMCID: PMC3194821 DOI: 10.1371/journal.pone.0026236] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 09/22/2011] [Indexed: 11/19/2022] Open
Abstract
Retinoic acid (RA) has been approved for the differentiation therapy of neuroblastoma (NB). Previous work revealed a correlation between glucose-regulated protein 75 (GRP75) and the RA-elicited neuronal differentiation of NB cells. The present study further demonstrated that GRP75 translocates into the nucleus and physically interacts with retinoid receptors (RARα and RXRα) to augment RA-elicited neuronal differentiation. GRP75 was required for RARα/RXRα-mediated transcriptional regulation and was shown to reduce the proteasome-mediated degradation of RARα/RXRαin a RA-dependent manner. More intriguingly, the level of GRP75/RARα/RXRα tripartite complexes was tightly associated with the RA-induced suppression of tumor growth in animals and the histological grade of differentiation in human NB tumors. The formation of GRP75/RARα/RXRα complexes was intimately correlated with a normal MYCN copy number of NB tumors, possibly implicating a favorable prognosis of NB tumors. The present findings reveal a novel function of nucleus-localized GRP75 in actively promoting neuronal differentiation, delineating the mode of action for the differentiation therapy of NB by RA.
Collapse
|
29
|
Lee M, Shin J. Triage of oxidation-prone proteins by Sqstm1/p62 within the mitochondria. Biochem Biophys Res Commun 2011; 413:122-7. [PMID: 21872578 DOI: 10.1016/j.bbrc.2011.08.067] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 08/13/2011] [Indexed: 11/16/2022]
Abstract
As the mitochondrion is vulnerable to oxidative stress, cells have evolved several strategies to maintain mitochondrial integrity, including mitochondrial protein quality control mechanisms and autophagic removal of damaged mitochondria. Involvement of an autophagy adaptor, Sqstm1/p62, in the latter process has been recently described. In the present study, we provide evidence that a portion of p62 directly localizes within the mitochondria and supports stable electron transport by forming heterogeneous protein complexes. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF) of mitochondrial proteins co-purified with p62 revealed that p62 interacts with several oxidation-prone proteins, including a few components of the electron transport chain complexes, as well as multiple chaperone molecules and redox regulatory enzymes. Accordingly, p62-deficient mitochondria exhibited compromised electron transport, and the compromised function was partially restored by in vitro delivery of p62. These results suggest that p62 plays an additional role in maintaining mitochondrial integrity at the vicinity of target machineries through its function in relation to protein quality control.
Collapse
Affiliation(s)
- Minjung Lee
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine and Samsung Biomedical Research Institute, Suwon-Si, Kyonggi-Do, Republic of Korea
| | | |
Collapse
|
30
|
Chen X, Xu B, Li H, Yang L, Zuo J, Liu W, Liu C. Expression of mortalin detected in human liver cancer by tissue microarrays. Anat Rec (Hoboken) 2011; 294:1344-51. [PMID: 21714113 DOI: 10.1002/ar.21433] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 05/03/2011] [Indexed: 12/12/2022]
Abstract
Mortalin is a highly conserved molecular chaperone in the heat shock protein (HSP) 70 family, which plays a role in carcinogenesis. The relationship between tumors and the expression of mortalin is not fully elucidated. In this study, human tumor specimens from various organs of liver cancer at different stages and cultured liver cancer cells were used to study the expression pattern of mortalin. Through immunohistochemistry we showed that mortalin was significantly higher in tumors than in adjacent benign tissues. Using liver tissue microarrays tested on hepatocellular carcinomas, mortalin expression was consecutively higher with advanced tumor stages. Mortalin expression on the cultured liver cancer cells were characterized with immunocytochemistry, Real-time PCR, and western blot. The results showed that the expression level is markedly higher in the SMMC 7721 (a liver-derived tumor cell line) than in the HL 7702 (a normal liver cell line) in the protein level only. Understanding the role of mortalin in tumors may lead to development of a new therapeutic target in cancer treatment.
Collapse
Affiliation(s)
- Xi Chen
- Department of Cellular and Genetic Medicine, Shanghai Medical College, Fudan University, China
| | | | | | | | | | | | | |
Collapse
|
31
|
Logan CA, Somero GN. Effects of thermal acclimation on transcriptional responses to acute heat stress in the eurythermal fish Gillichthys mirabilis (Cooper). Am J Physiol Regul Integr Comp Physiol 2011; 300:R1373-83. [DOI: 10.1152/ajpregu.00689.2010] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The capacities of eurythermal ectotherms to withstand wide ranges of temperature are based, in part, on abilities to modulate gene expression as body temperature changes, notably genes encoding proteins of the cellular stress response. Here, using a complementary DNA microarray, we investigated the sequence in which cellular stress response-linked genes are expressed during acute heat stress, to elucidate how severity of stress affects the categories of genes changing expression. We also studied how prior acclimation history affected gene expression in response to acute heat stress. Eurythermal goby fish ( Gillichthys mirabilis ) were acclimated to 9 ± 0.5, 19 ± 0.5, and 28 ± 0.5°C for 1 mo. Then fish were given an acute heat ramp (4°C/h), and gill tissues were sampled every +4°C to monitor gene expression. The average onset temperature for a significant change in expression during acute stress increased by ∼2°C for each ∼10°C increase in acclimation temperature. For some genes, warm acclimation appeared to obviate the need for expression change until the most extreme temperatures were reached. Sequential expression of different categories of genes reflected severity of stress. Regardless of acclimation temperature, the gene encoding heat shock protein 70 ( HSP70) was upregulated strongly during mild stress; the gene encoding the proteolytic protein ubiquitin ( UBIQ) was upregulated at slightly higher temperatures; and a gene encoding a protein involved in cell cycle arrest and apoptosis, cyclin-dependent kinase inhibitor 1B ( CDKN1B), was upregulated only under extreme stress. The tiered, stress level-related expression patterns and the effects of acclimation on induction temperature yield new insights into the fundamental mechanisms of eurythermy.
Collapse
Affiliation(s)
- Cheryl A. Logan
- Hopkins Marine Station, Department of Biology, Stanford University, Pacific Grove, California
| | - George N. Somero
- Hopkins Marine Station, Department of Biology, Stanford University, Pacific Grove, California
| |
Collapse
|
32
|
Lim Y, Hong E, Kwon D, Lee E. Proteomic identification and comparative analysis of asymmetrically arginine-methylated proteins in immortalized, young and senescent cells. Electrophoresis 2011; 31:3823-33. [PMID: 21080485 DOI: 10.1002/elps.201000361] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Protein-arginine methylation is one of the modifications that yields mono and dimethyl (asymmetric or symmetric) arginine residues in proteins. Previously, we found that asymmetric arginine methylation is decreased proportionately with a decrease of cell proliferation potential of cells, and such arginine methylation is greatest in immortalized cells, followed by normal young cells, and lowest in replicatively senescent cells. Using an asymmetric dimethyl-arginine-specific antibody, we identified arginine-methylated proteins in these cell types by immunoprecipitation and 2-D immunoblotting followed by MS. As a result, arginine methylation of chaperone molecules and RNA-binding proteins was differentially regulated between immortalized or young cells and senescent cells. Immortalized cells had significantly higher levels of methyl-accepting proteins, such as cleavage stimulation factor 2 (CstF2) and heterogeneous nuclear ribonucleoprotein (hnRNP) R, than young cells. However, senescent cells contained hypomethylated CstF2, hnRNP K, and chaperone containing TCP1 subunit 7, as well as decreased hnRNP R level. Further, significant reduction of arginine modification in CstF2 and chaperone containing TCP1 subunit 7 was observed in prematurely senescent fibroblasts, induced by treatment with adenosine dialdehyde, a transmethylation inhibitor, or subcytotoxic concentration of H(2)O(2). These results suggest that asymmetric modification of RNA-binding proteins and molecular chaperones plays an essential role in maintaining cell proliferation capability.
Collapse
Affiliation(s)
- Yongchul Lim
- Cellular and Developmental Biology, Division of Brain Korea 21 Program for Biomedical Sciences, Korea University, Seoul, Korea
| | | | | | | |
Collapse
|
33
|
Luo WI, Dizin E, Yoon T, Cowan JA. Kinetic and structural characterization of human mortalin. Protein Expr Purif 2010; 72:75-81. [PMID: 20152901 DOI: 10.1016/j.pep.2010.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Revised: 02/04/2010] [Accepted: 02/08/2010] [Indexed: 11/25/2022]
Abstract
Human mortalin is an Hsp70 chaperone that has been implicated in cancer, Alzheimer's and Parkinson's disease, and involvement has been suggested in cellular iron-sulfur cluster biosynthesis. However, study of this important human chaperone has been hampered by a lack of active material sufficient for biochemical characterization. Herein, we report the successful purification and characterization of recombinant human mortalin in Escherichia coli. The recombinant protein was expressed in the form of inclusion bodies and purified by Ni-NTA affinity chromatography. The subsequently refolded protein was confirmed to be active by its ATPase activity, a characteristic blue-shift in the fluorescence emission maximum following the addition of ATP, and its ability to bind to a likely physiological substrate. Single turnover kinetic experiments of mortalin were performed and compared with another Hsp70 chaperone, Thermotogamaritima DnaK; with each exhibiting slow ATP turnover rates. Secondary structures for both chaperones were similar by circular dichroism criteria. This work describes an approach to functional expression of human mortalin that provides sufficient material for detailed structure-function studies of this important Hsp70 chaperone.
Collapse
Affiliation(s)
- Wen-I Luo
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, United States
| | | | | | | |
Collapse
|
34
|
Yi H, Li XH, Yi B, Zheng J, Zhu G, Li C, Li MY, Zhang PF, Li JL, Chen ZC, Xiao ZQ. Identification of Rack1, EF-Tu and Rhodanese as Aging-Related Proteins in Human Colonic Epithelium by Proteomic Analysis. J Proteome Res 2010; 9:1416-23. [PMID: 20099848 DOI: 10.1021/pr9009386] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hong Yi
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xin-Hui Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Bin Yi
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Zheng
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Guo Zhu
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Cui Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Mao-Yu Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Peng-Fei Zhang
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jian-Ling Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhu-Chu Chen
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhi-Qiang Xiao
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
35
|
Proteomic analysis of regenerating mouse liver following 50% partial hepatectomy. Proteome Sci 2009; 7:48. [PMID: 20040084 PMCID: PMC2813229 DOI: 10.1186/1477-5956-7-48] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Accepted: 12/29/2009] [Indexed: 12/29/2022] Open
Abstract
Background Although 70% (or 2/3) partial hepatectomy (PH) is the most studied model for liver regeneration, the hepatic protein expression profile associated with lower volume liver resection (such as 50% PH) has not yet been reported. Therefore, the aim of this study was to determine the global protein expression profile of the regenerating mouse liver following 50% PH by differential proteomics, and thereby gaining some insights into the hepatic regeneration mechanism(s) under this milder but clinically more relevant condition. Results Proteins from sham-operated mouse livers and livers regenerating for 24 h after 50% PH were separated by SDS-PAGE and analyzed by nanoUPLC-Q-Tof mass spectrometry. Compared to sham-operated group, there were totally 87 differentially expressed proteins (with 50 up-regulated and 37 down-regulated ones) identified in the regenerating mouse livers, most of which have not been previously related to liver regeneration. Remarkably, over 25 differentially expressed proteins were located at mitochondria. Several of the mitochondria-resident proteins which play important roles in citric acid cycle, oxidative phosphorylation and ATP production were found to be down-regulated, consistent with the recently-proposed model in which the reduction of ATP content in the remnant liver gives rise to early stress signals that contribute to the onset of liver regeneration. Pathway analysis revealed a central role of c-Myc in the regulation of liver regeneration. Conclusions Our study provides novel evidence for mitochondria as a pivotal organelle that is connected to liver regeneration, and lays the foundation for further studies on key factors and pathways involved in liver regeneration following 50% PH, a condition frequently used for partial liver transplantation and conservative liver resection.
Collapse
|
36
|
Gabriele J, Pontoriero GF, Thomas N, Thomson CA, Skoblenick K, Pristupa ZB, Mishra RK. Cloning, characterization, and functional studies of a human 40-kDa catecholamine-regulated protein: implications in central nervous system disorders. Cell Stress Chaperones 2009; 14:555-67. [PMID: 19280369 PMCID: PMC2866950 DOI: 10.1007/s12192-009-0107-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 02/09/2009] [Accepted: 02/19/2009] [Indexed: 12/14/2022] Open
Abstract
Catecholamine-regulated proteins (CRPs) have been shown to bind dopamine and other structurally related catecholamines; in particular, the 40-kDa CRP (CRP40) protein has been previously cloned and functionally characterized. To determine putative human homologs, BLAST analysis using the bovine CRP40 sequence identified a human established sequence tag (EST) with significant homology (accession #BQ224193). Using this EST, we cloned a recombinant human brain CRP40-like protein, which possessed chaperone activity. Radiolabeled dopamine binding studies with recombinant human CRP40 protein demonstrated the ability of this protein to bind dopamine with low affinity and high capacity. The full-length human CRP40 nucleotide sequence was elucidated (accession #DQ480334) with RNA ligase-mediated rapid amplification of complementary DNA ends polymerase chain reaction, while Northern blot hybridization suggested that human CRP40 is an alternative splice variant of the 70-kDa mitochondrial heat shock protein, mortalin. Human SH-SY5Y neuroblastoma cells treated with the antipsychotic drug, haloperidol, exhibited a significant increase in CRP40 messenger RNA expression compared to untreated control cells, while other dopamine agonists/antagonists also altered CRP40 expression and immunolocalization. In conclusion, these results show that we have cloned a splice variant of mortalin with a novel catecholamine binding function and that this chaperone-like protein may be neuroprotective in dopamine-related central nervous system disorders.
Collapse
Affiliation(s)
- Joseph Gabriele
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
| | | | | | | | | | | | | |
Collapse
|
37
|
Conte M, Deri P, Isolani ME, Mannini L, Batistoni R. A mortalin-like gene is crucial for planarian stem cell viability. Dev Biol 2009; 334:109-18. [PMID: 19616535 DOI: 10.1016/j.ydbio.2009.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 07/08/2009] [Accepted: 07/09/2009] [Indexed: 12/22/2022]
Abstract
In adult organisms, stem cells are crucial to homeostasis and regeneration of damaged tissues. In planarians, adult stem cells (neoblasts) are endowed with an extraordinary replicative potential that guarantees unlimited replacement of all differentiated cell types and extraordinary regenerative ability. The molecular mechanisms by which neoblasts combine long-term stability and constant proliferative activity, overcoming the impact of time, remain by far unknown. Here we investigate the role of Djmot, a planarian orthologue that encodes a peculiar member of the HSP70 family, named Mortalin, on the dynamics of stem cells of Dugesia japonica. Planarian stem cells and progenitors constitutively express Djmot. Transient Djmot expression in differentiated tissues is only observed after X-ray irradiation. DjmotRNA interference causes inability to regenerate and death of the animals, as a result of permanent growth arrest of stem cells. These results provide the first evidence that an hsp-related gene is essential for neoblast viability and suggest the possibility that high levels of Djmot serve to keep a p53-like protein signaling under control, thus allowing neoblasts to escape cell death programs. Further studies are needed to unravel the molecular pathways involved in these processes.
Collapse
Affiliation(s)
- Maria Conte
- Dipartimento di Biologia, Università di Pisa, I-56017 Ghezzano, Italy
| | | | | | | | | |
Collapse
|
38
|
Sowell RA, Owen JB, Butterfield DA. Proteomics in animal models of Alzheimer's and Parkinson's diseases. Ageing Res Rev 2009; 8:1-17. [PMID: 18703168 DOI: 10.1016/j.arr.2008.07.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 07/04/2008] [Accepted: 07/08/2008] [Indexed: 01/06/2023]
Abstract
The risk of developing neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD) increases with age. AD and PD are the two most common neurodegenerative diseases that currently affect millions of persons within the United States population. While many clues about the mechanisms of these disorders have been uncovered, to date, the molecular mechanisms associated with the cause of these diseases are not completely understood. Furthermore, there are no available cures or preventive treatments for either disorder. Animal models of AD and PD, though not perfect, offer a means to gain knowledge of the basic biochemistry associated with these disorders and with drug efficacy. The field of proteomics which focuses on identifying the dynamic nature of the protein content expressed within a particular cell, tissue, or organism, has provided many insights into these disturbing disorders. Proteomic studies have revealed many pathways that are associated with disease pathogenesis and that may lead to the development of potential therapeutic targets. This review provides a discussion of key findings from AD and PD proteomics-based studies in various animal models of disease.
Collapse
Affiliation(s)
- Renã A Sowell
- Department of Chemistry, University of Kentucky, Lexington, KY 40506-0055, USA
| | | | | |
Collapse
|
39
|
Wang M, Windgassen D, Papoutsakis ET. A global transcriptional view of apoptosis in human T-cell activation. BMC Med Genomics 2008; 1:53. [PMID: 18947405 PMCID: PMC2600644 DOI: 10.1186/1755-8794-1-53] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Accepted: 10/23/2008] [Indexed: 12/22/2022] Open
Abstract
Background T-cell activation is an essential step of immune response. The process of proper T-cell activation is strictly monitored and regulated by apoptosis signaling. Yet, regulation of apoptosis, an integral and crucial facet during the process of T-cell activation, is not well understood. Methods In this study, a Gene-Ontology driven global gene expression analysis coupled with protein abundance and activity assays identified genes and pathways associated with regulation of apoptosis in primary human CD3+ T cells and separately CD4+ and CD8+ T cells. Results We identified significantly regulated apoptotic genes in several protein families, such as BCL2 proteins, CASPASE proteins, and TNF receptors, and detailed their transcriptional kinetics during the T-cell activation process. Transcriptional patterns of a few select genes (BCL2A1, BBC3 and CASP3) were validated at the protein level. Many of these apoptotic genes are involved in NF-κB signaling pathway, including TNFRSF10A, TNFRSF10B, TRAF4, TRAF1, TRAF3, and TRAF6. Upregulation of NF-κB and IκB family genes (REL, RELA, and RELB, NFKBIA, NFKBIE and NFKB1) at 48 to 96 hours, supported by the increase of phosphorylated RELA (p65), suggests that the involvement of the NF-κB complex in the process of T-cell proliferation is not only regulated at the protein level but also at the transcriptional level. Examination of genes involved in MAP kinase signalling pathway, important in apoptosis, suggests an induction of p38 and ERK1 cascades in T-cell proliferation (at 48 to 96 hours), which was explored using phosphorylation assays for p38 (MAPK14) and ERK1 (MAPK3). An immediate and short-lived increase of AP-1 activity measured by DNA-binding activity suggests a rapid and transient activation of p38 and/or JNK cascades upon T-cell activation. Conclusion This comparative genome-scale, transcriptional analysis of T-cell activation in the CD4+ and CD8+ subsets and the mixed CD3+ population identified many apoptosis genes not previously identified in the context of T-cell activation. Furthermore, it provided a comprehensive temporal analysis of the transcriptional program of apoptosis associated with T-cell activation.
Collapse
Affiliation(s)
- Min Wang
- Interdepartmental Biological Sciences Program, Northwestern University, Evanston, IL, USA.
| | | | | |
Collapse
|
40
|
Jiang P, Siggers JLA, Ngai HHY, Sit WH, Sangild PT, Wan JMF. The small intestine proteome is changed in preterm pigs developing necrotizing enterocolitis in response to formula feeding. J Nutr 2008; 138:1895-901. [PMID: 18806098 DOI: 10.1093/jn/138.10.1895] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in newborn premature infants. Clinical studies show increased incidence of NEC in premature infants with enteral formula feeding; however, pathogenesis remains unclear. To identify the NEC-related proteins for molecular mechanisms, we applied proteomics analysis to characterize changes in the protein expression profile of newborn premature piglet intestines with NEC developed after enteral formula feeding for 24 h. Changes in protein expression were identified using 2-dimensional gel electrophoresis and peptide mass fingerprinting with MS as well as western blotting analysis. Nineteen differentially expressed proteins were identified and these have roles in oxidative stress, chaperone, signal transduction, protein folding and degradation, oxygen transport, signal transduction, and energy metabolism. Proteins with increased levels include manganese-containing superoxide dismutase and hemoglobin subunit and proteins with decreased expression include sorbitol dehydrogenase, mitochondrial aldehyde dehydrogenase 2, glucose-regulated protein 75, CRY protein, snail homolog 3, thyroid hormone-binding protein precursor, and DJ1 (Parkinson's disease 7) etc. The data provided novel mechanistic insights into the pathogenesis of NEC and the insults of a formulated diet to the premature gut.
Collapse
Affiliation(s)
- Pingping Jiang
- Division of Agricultural, Food and Nutritional Science, School of Biological Sciences, The University of Hong Kong, Hong Kong S.A.R., P.R. China
| | | | | | | | | | | |
Collapse
|
41
|
Hsu WM, Lee H, Juan HF, Shih YY, Wang BJ, Pan CY, Jeng YM, Chang HH, Lu MY, Lin KH, Lai HS, Chen WJ, Tsay YG, Liao YF, Hsieh FJ. Identification of GRP75 as an Independent Favorable Prognostic Marker of Neuroblastoma by a Proteomics Analysis. Clin Cancer Res 2008; 14:6237-45. [DOI: 10.1158/1078-0432.ccr-07-4181] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
42
|
Deocaris CC, Takano S, Priyandoko D, Kaul Z, Yaguchi T, Kraft DC, Yamasaki K, Kaul SC, Wadhwa R. Glycerol stimulates innate chaperoning, proteasomal and stress-resistance functions: implications for geronto-manipulation. Biogerontology 2008; 9:269-82. [DOI: 10.1007/s10522-008-9136-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Accepted: 02/27/2008] [Indexed: 12/21/2022]
|
43
|
Bermejo-Nogales A, Benedito-Palos L, Saera-Vila A, Calduch-Giner JA, Sitjà-Bobadilla A, Pérez-Sánchez J. Confinement exposure induces glucose regulated protein 75 (GRP75/mortalin/mtHsp70/PBP74/HSPA9B) in the hepatic tissue of gilthead sea bream (Sparus aurata L.). Comp Biochem Physiol B Biochem Mol Biol 2008; 149:428-38. [DOI: 10.1016/j.cbpb.2007.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2007] [Revised: 11/13/2007] [Accepted: 11/14/2007] [Indexed: 11/25/2022]
|
44
|
Kaul Z, Yaguchi T, Harada JI, Ikeda Y, Hirano T, Chiura HX, Kaul SC, Wadhwa R. An antibody-conjugated internalizing quantum dot suitable for long-term live imaging of cells. Biochem Cell Biol 2007; 85:133-40. [PMID: 17464353 DOI: 10.1139/o06-205] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Quantum dots (QD) are fluorescent semiconductor nanocrystals that are emerging as superior alternatives to the conventional organic dyes used in biological applications. Although QDs offer several advantages over conventional fluorescent dyes, including greater photostability and a wider range of excitation and (or) emission wavelengths, their toxicity has been an issue in its wider use as an analytic, diagnostic and therapeutic tool. We prepared a conjugate QD with an internalizing antibody and demonstrated that the QD-antibody conjugate is efficiently internalized into cells and is visible even after multiple divisions. We demonstrate that the internalized QD is nontoxic to cells and provides a sensitive tool for long-term molecular imaging.
Collapse
Affiliation(s)
- Zeenia Kaul
- International Christian University, Division of Natural Sciences, 3-10-2 Osawa, Mitaka, Tokyo 181-8585, Japan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Zvereff V, Wang JC, Shun K, Lacoste J, Chevrette M. Colocalisation of CD9 and mortalin in CD9-induced mitotic catastrophe in human prostate cancer cells. Br J Cancer 2007; 97:941-8. [PMID: 17848953 PMCID: PMC2360413 DOI: 10.1038/sj.bjc.6603964] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
CD9, a member of the tetraspanin family of proteins, is involved in a variety of cellular interactions with many other proteins and molecules. Although CD9 has been implicated in cell fusion, migration and cancer progression, the detailed function of this protein is not completely understood and likely depends on interactions with different protein partners, which are not yet all known. Using co-immunoprecipitation and mass-spectrometric protein sequencing, we have identified in prostate cancer cells, a novel CD9 partner, the 75-kDa protein HSPA9B, also known as mortalin. We further show that introduction and overexpression of wild-type CD9 into human PC-3 prostate cancer cells induces mitotic catastrophe. We also demonstrate, by immunocolocalisation studies, the interaction of CD9 and mortalin in PC-3 cells undergoing mitotic catastrophe. Our results not only identified mortalin as a new CD9 partner, but also clarify the mechanisms by which CD9 may control prostate cancer progression.
Collapse
Affiliation(s)
- V Zvereff
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - J-C Wang
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - K Shun
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - J Lacoste
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal Quebec, Canada
| | - M Chevrette
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- The Research Institute of the McGill University Health Centre, 1650 Cedar Ave, Room R4-113, Montreal, Quebec, Canada H3G 1A4. E-mail:
| |
Collapse
|
46
|
Ohtsuka R, Abe Y, Fujii T, Yamamoto M, Nishimura J, Takayanagi R, Muta K. Mortalin is a novel mediator of erythropoietin signaling. Eur J Haematol 2007; 79:114-25. [PMID: 17635236 DOI: 10.1111/j.1600-0609.2007.00870.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Erythropoietin (EPO) stimulates erythroid growth by enhancing the proliferation, maturation and survival of late-stage erythroid progenitor cells. However, the entire process of EPO stimulation remains undetermined. To further clarify the intracellular mechanisms by which EPO affects the growth of erythroid progenitor cells, we analyzed proteins obtained from purified human erythroid colony-forming cells (ECFCs) cultured with or without EPO, and one of the proteins apparently related with EPO stimuli was identified as mortalin (mthsp70/PBP74/Grp75/mot-2), which is a member of the heat shock protein 70 family of chaperones. The amount of mortalin mRNA in ECFCs increased in an EPO dose-dependent manner, and ECFC growth was dependent on the amount of mortalin. Furthermore, expression of mortalin in ECFCs was suppressed by a phosphatidylinositol 3-kinase inhibitor. Finally, we analyzed gene expression patterns in ECFCs cultured with or without EPO after treatment with mortalin small interfering RNA (siRNA) using a DNA microarray. When ECFCs treated with mortalin siRNA were cultured with EPO, the expression of several genes overlapped with the profile seen in control ECFCs cultured without EPO. Our data suggest that mortalin is involved in the mediation of EPO signaling and plays an important role in stimulating the growth of erythroid progenitor cells.
Collapse
Affiliation(s)
- Rie Ohtsuka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Kanai M, Ma Z, Izumi H, Kim SH, Mattison CP, Winey M, Fukasawa K. Physical and functional interaction between mortalin and Mps1 kinase. Genes Cells 2007; 12:797-810. [PMID: 17573779 DOI: 10.1111/j.1365-2443.2007.01091.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mortalin is a member of Hsp70 chaperoning protein family involved in various cellular functions. Through the search of the kinases that mortalin physically interact with, we identified Mps1 as such a kinase. Mps1 kinase has been implicated in the regulation of centrosome duplication and mitotic checkpoint response. Mortalin binds to Mps1, and is phosphorylated by Mps1 on Thr62 and Ser65. The phosphorylated mortalin then super-activates Mps1 in a feedback manner. Mortalin has been previously shown to localize to centrosomes, and to be involved in the regulation of centrosome duplication. We found that centrosomal localization of mortalin depends on the presence of Mps1. Moreover, Mps1-associated acceleration of centrosome duplication depends on the presence of mortalin and super-activation by the Thr62/Ser65 phosphorylated mortalin.
Collapse
Affiliation(s)
- Masayuki Kanai
- Department of Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Korrapati MC, Chilakapati J, Witzmann FA, Rao C, Lock EA, Mehendale HM. Proteomics of S-(1, 2-dichlorovinyl)-L-cysteine-induced acute renal failure and autoprotection in mice. Am J Physiol Renal Physiol 2007; 293:F994-F1006. [PMID: 17581926 DOI: 10.1152/ajprenal.00114.2007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous studies (Vaidya VS, Shankar K, Lock EA, Bucci TJ, Mehendale HM. Toxicol Sci 74: 215-227, 2003; Korrapati MC, Lock EA, Mehendale HM. Am J Physiol Renal Physiol 289: F175-F185, 2005; Korrapati MC, Chilakapati J, Lock EA, Latendresse JR, Warbritton A, Mehendale HM. Am J Physiol Renal Physiol 291: F439-F455, 2006) demonstrated that renal repair stimulated by a low dose of S-(1,2-dichlorovinyl)l-cysteine (DCVC; 15 mg/kg i.p.) 72 h before administration of a normally lethal dose (75 mg/kg i.p.) protects mice from acute renal failure (ARF) and death (autoprotection). The present study identified the proteins indicative of DCVC-induced ARF and autoprotection in male Swiss Webster mice. Renal dysfunction and injury were assessed by plasma creatinine and histopathology, respectively. Whole-kidney homogenates were run on two-dimensional gel electrophoresis gels, and the expression of 18 common proteins was maximally changed (> or =10-fold) in all the treatment groups and they were conclusively identified by liquid chromatography tandem mass spectrometry. These proteins were mildly downregulated after low dose alone and in autoprotected mice in contrast to severe downregulation with high dose alone. Glucose-regulated protein 75 and proteasome alpha-subunit type 1 were further investigated by immunohistochemistry for their localization in the kidneys of all the groups. These proteins were substantially higher in the proximal convoluted tubular epithelial cells in the low-dose and autoprotected groups compared with high-dose alone group. Proteins involved in energetics were downregulated in all the three groups of mice, leading to a compromise in cellular energy. However, energy is recovered completely in low-dose and autoprotected mice. This study provides the first report on proteomics of DCVC-induced ARF and autoprotection in mice and reflects the application of proteomics in mechanistic studies as well as biomarker development in a variety of toxicological paradigms.
Collapse
Affiliation(s)
- Midhun C Korrapati
- Department of Toxicology, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71209-0470, USA
| | | | | | | | | | | |
Collapse
|
49
|
Vaishnav RA, Getchell ML, Poon HF, Barnett KR, Hunter SA, Pierce WM, Klein JB, Butterfield DA, Getchell TV. Oxidative stress in the aging murine olfactory bulb: redox proteomics and cellular localization. J Neurosci Res 2007; 85:373-85. [PMID: 17131389 DOI: 10.1002/jnr.21130] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A recent proteomics analysis from our laboratory demonstrated that several oxidative stress response proteins showed significant changes in steady-state levels in olfactory bulbs (OBs) of 20- vs. 1.5-month-old mice. Oxidative stress may result in protein oxidation. In this study, we investigated two forms of protein oxidative modification in murine OBs: carbonylation and nitration. Redox proteomics with two-dimensional gel electrophoresis, Western blotting, protein digestion, and mass spectrometry was used to quantify total and specific protein carbonylation and to identify differentially carbonylated proteins and determine the carbonylation status of previously identified proteins in OBs of 1.5- and 20-month-old mice. Immunohistochemistry was used to demonstrate the relative intensity and localization of protein nitration in OBs of 1.5-, 6-, and 20-month-old mice. Total protein carbonylation was significantly greater in OBs of 20- vs. 1.5-month-old mice. Aldolase 1 (ALDO1) showed significantly more carbonylation in OBs from 20- vs. 1.5-month-old mice; heat shock protein 9A and dihydropyrimidinase-like 2 showed significantly less. Several previously investigated proteins were also carbonylated, including ferritin heavy chain (FTH). Nitration, identified by 3-nitrotyrosine immunoreactivity, was least abundant at 1.5 months, intermediate at 6 months, and greatest at 20 months and was localized primarily in blood vessels. Proteins that were specific targets of oxidation were also localized: ALDO1 in astrocytes of the granule cell layer and FTH in mitral/tufted cells. These results indicate that specific carbonylated proteins, including those in astrocytes and mitral/tufted neurons, and nitrated proteins in the vasculature are molecular substrates of age-related olfactory dysfunction.
Collapse
Affiliation(s)
- Radhika A Vaishnav
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Alvares K, Dixit SN, Lux E, Barss J, Veis A. The proteome of the developing tooth of the sea urchin,Lytechinus variegatus: mortalin is a constituent of the developing cell syncytium. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2007; 308:357-70. [PMID: 17385701 DOI: 10.1002/jez.b.21159] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Echinoderm teeth are continuously growing calcite-mineralized tissues of complex structure. Two features are of special interest: (1) cell division takes place in a restricted aboral domain, the plumula, and the cells immediately merge into multinucleated syncytial layers; (2) the major part of the heavily mineralized tooth elongates and moves towards the adoral incisal tip continuously as the syncytial cells actively expand the syncytium and intermembrane mineral phase. As the first step to understanding the nature of the mineralization processes, we have isolated the proteins of the plumula and of the mature mineralized portions of the tooth, and begun their characterization. Peptide sequences were used to screen a plumula cDNA library by polymerase chain reaction. One primer set yielded a prominent amplified product which was cloned, and sequenced. Comparison with the nucleotide and protein data banks revealed the protein to be Mortalin, a member of the hsp-70 family, with >75% of its sequences identical to that of human mortalin. Immunocytochemical localization of mortalin within the plumula, using Anti-human Grp75, showed staining of the odontoblast cytosol and matrix at the point where syncytial formation was occurring. The cytosol of the syncytial layers was weakly stained. The nuclei within the syncytia were stained at their periphery. In the mature part of the tooth, the perinuclear staining of the nuclei was more prominent. We conclude that mortalin is involved in syncytium formation and maintenance. The urchin mortalin has a distinctive aspartic acid and serine-rich C-terminal domain that may link it to the mineralization process.
Collapse
Affiliation(s)
- Keith Alvares
- Department of Cell and Molecular Biology, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|