1
|
Huang Y, Zhao Y, Liu H, Yang Y, Cheng L, Deng X, Chao L. Decreased expression of GRIM-19 induces autophagy through the AMPK/ULK1 signaling pathway during adenomyosis. Biol Reprod 2022; 107:956-966. [PMID: 35908189 DOI: 10.1093/biolre/ioac151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 06/07/2022] [Indexed: 11/14/2022] Open
Abstract
The processes underlying adenomyosis are similar to those of tumor metastasis, and it is defined as progressive invasion by the endometrium and the subsequent creation of ectopic lesions. GRIM-19 regulates cell death via the mitochondrial respiratory chain. Stress following oxygen deprivation can induce tumor cell autophagy, leading to cell invasion and migration. Here, we revealed that GRIM-19 negatively regulates autophagy, and, at least in adenomyosis, decreased expression of GRIM-19 is accompanied by an increased level of autophagy and 5'-adenosine monophosphate-activated protein kinase-Unc-51 like autophagy activating kinase 1 (AMPK-ULK1) activation. Upregulation of GRIM-19 expression in human primary endometrial cells and ISHIKAWA cells inhibits autophagy via the AMPK-ULK1 pathway and helps control cell invasion and migration. In addition, we also identified increased expression of AMPK and ULK1, and higher levels of autophagy in the uterine tissues of GRIM-19+/- mice. Importantly, the function of the GRIM-19-AMPK-ULK1 axis in regulating autophagy in adenomyosis is similar to that of tumor tissues, which may help elucidate the regulation of adenomyosis tumor-like behavior, and is expected to help identify novel targets for the diagnosis and treatment of adenomyosis.
Collapse
Affiliation(s)
- YuFei Huang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Yue Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - HaoRan Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Yang Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - LaiYang Cheng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - XiaoHui Deng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Lan Chao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| |
Collapse
|
2
|
Nallar SC, Kalvakolanu DV. GRIM-19: A master regulator of cytokine induced tumor suppression, metastasis and energy metabolism. Cytokine Growth Factor Rev 2016; 33:1-18. [PMID: 27659873 DOI: 10.1016/j.cytogfr.2016.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 09/14/2016] [Indexed: 12/31/2022]
Abstract
Cytokines induce cell proliferation or growth suppression depending on the context. It is increasingly becoming clear that success of standard radiotherapy and/or chemotherapeutics to eradicate solid tumors is dependent on IFN signaling. In this review we discuss the molecular mechanisms of tumor growth suppression by a gene product isolated in our laboratory using a genome-wide expression knock-down strategy. Gene associated with retinoid-IFN-induced mortality -19 (GRIM-19) functions as non-canonical tumor suppressor by antagonizing oncoproteins. As a component of mitochondrial respiratory chain, GRIM-19 influences the degree of "Warburg effect" in cancer cells as many advanced and/or aggressive tumors show severely down-regulated GRIM-19 levels. In addition, GRIM-19 appears to regulate innate and acquired immune responses in mouse models. Thus, GRIM-19 is positioned at nodes that favor cell protection and/or prevent aberrant cell growth.
Collapse
Affiliation(s)
- Shreeram C Nallar
- Department of Microbiology and Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dhan V Kalvakolanu
- Department of Microbiology and Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
3
|
Chen L, Cui H, Fang J, Deng H, Kuang P, Guo H, Wang X, Zhao L. Glutamine deprivation plus BPTES alters etoposide- and cisplatin-induced apoptosis in triple negative breast cancer cells. Oncotarget 2016; 7:54691-54701. [PMID: 27419628 PMCID: PMC5342373 DOI: 10.18632/oncotarget.10579] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/30/2016] [Indexed: 12/25/2022] Open
Abstract
Glutamine provides cancer cells with the energy required to synthesize macromolecules. Methods which block glutamine metabolism in treatment of breast cancer inhibit oncogenic transformation and tumor growth. We investigated whether inhibiting glutamine metabolism produces effects that are synergistic with those produced by drugs which damage DNA in triple-negative breast cancer cells. HCC1937 and BT-549 breast cancer cells were co-treated with either cisplatin or etoposide in combination with BPTES (a specific inhibitor of glutaminase 1) or exposure to a glutamine-free medium, and the cell proliferation and cell apoptosis were measured by flow cytometry, immunoblotting studies, and CCK-8 assays. The results showed that both glutamine deprivation and BPTES pretreatments increased the toxic effects of cisplatin and etoposide on HCC1937 cells, as demonstrated by their reduced proliferation, increased expression of apoptosis-related proteins (cleaved-PARP, cleaved-caspase 9, and cleaved-caspase 3) and decreased Bcl-2/BAX ratio. However, in BT-549 cells, glutamine deprivation and BPTES treatment increased etoposide-induced apoptosis only when used with higher concentrations of etoposide, and the effect on cisplatin-induced apoptosis was minimal. These results suggest that the anti-cancer effects produced by a combined approach of inhibiting glutamine metabolism and administering common chemotherapeutic agents correlate with the tumor cell type and specific drugs being administered.
Collapse
Affiliation(s)
- Lian Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an 625014, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an 625014, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Ya'an 625014, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an 625014, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Ya'an 625014, China
| | - Huidan Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an 625014, China
| | - Ping Kuang
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an 625014, China
| | - Hongrui Guo
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an 625014, China
| | - Xun Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an 625014, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Ya'an 625014, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an 625014, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Ya'an 625014, China
| |
Collapse
|
4
|
Kong D, Zhao L, Du Y, He P, Zou Y, Yang L, Sun L, Wang H, Xu D, Meng X, Sun X. Overexpression of GRIM-19, a mitochondrial respiratory chain complex I protein, suppresses hepatocellular carcinoma growth. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:7497-7507. [PMID: 25550785 PMCID: PMC4270598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 10/16/2014] [Indexed: 06/04/2023]
Abstract
GRIM-19 has been demonstrated as an important regulator for the normal tissue development. Recently, more evidences regarded GRIM-19 as the new tumor suppressor. However, the possible mechanisms underlying GRIM-19 suppressing cancer growth are unclear. In the present study, Paired hepatocellular carcinoma (HCC) and adjacent non-tumor liver tissues were obtained from 54 patients who underwent primary surgical HCC tissue resection. GRIM-19 protein expression in HCC tissues was performed by immunohistochemistry. Cells were transfected by lentiviruses plasmid expressing GRIM-19. RT-PCR and Western blot analyses were performed to confirm the expression of GRIM-19 mRNA or protein. Cell proliferation was assessed by MTT and FCM analyses. Mitochondrial membrane potential and apoptosis were respectively determined by using fluorescence microscopy and FCM analyses. AKT1, pAKT1, cyclinD1, CDK4, PCNA, Bax, Bcl-2, cleaved caspase-9, cleaved caspase-3, and cytochrome C were detected by Western blot and immunofluorescence. GRIM-19 protein expression was markedly lower in HCC than in paired adjacent non-tumor liver tissues. GRIM-19 overexpression in HCC cells significantly induced cell cycle arrest and enhanced apoptosis. We also found that AKT1 expression and phosphorylation were regulated by the expression of GRIM-19. Collectively, our study demonstrated that GRIM-19 overexpression suppressed HCC growth and downregulated AKT1 expression, suggesting that GRIM-19 might play a crucial role in hepatocarcinogenesis through negatively regulating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Dexia Kong
- Department of Gastroenterology, First Hospital of Jilin UniversityChangchun 130021, China
- Department of Pathophysiology, School of Basic Medicine, Jilin UniversityChangchun 130021, China
| | - Lijing Zhao
- Department of Pathophysiology, School of Basic Medicine, Jilin UniversityChangchun 130021, China
| | - Yanwei Du
- Department of Pathophysiology, School of Basic Medicine, Jilin UniversityChangchun 130021, China
| | - Ping He
- Department of Gastroenterology, First Hospital of Jilin UniversityChangchun 130021, China
| | - Yabin Zou
- Department of Pathology, First Hospital of Jilin UniversityChangchun 130021, China
| | - Luoluo Yang
- Department of Gastroenterology, First Hospital of Jilin UniversityChangchun 130021, China
| | - Liankun Sun
- Department of Pathophysiology, School of Basic Medicine, Jilin UniversityChangchun 130021, China
| | - Hebin Wang
- Department of Pathophysiology, School of Basic Medicine, Jilin UniversityChangchun 130021, China
| | - Deqi Xu
- New Vaccine National Engineering Research Center, Beijing Three-Room South HospitalBeijing 100024, China
| | - Xiangwei Meng
- Department of Gastroenterology, First Hospital of Jilin UniversityChangchun 130021, China
| | - Xun Sun
- Department of Pathology, First Hospital of Jilin UniversityChangchun 130021, China
| |
Collapse
|
5
|
Guha P, Bandyopadhyaya G, Polumuri SK, Chumsri S, Gade P, Kalvakolanu DV, Ahmed H. Nicotine promotes apoptosis resistance of breast cancer cells and enrichment of side population cells with cancer stem cell-like properties via a signaling cascade involving galectin-3, α9 nicotinic acetylcholine receptor and STAT3. Breast Cancer Res Treat 2014; 145:5-22. [PMID: 24668500 DOI: 10.1007/s10549-014-2912-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 03/13/2014] [Indexed: 10/25/2022]
Abstract
Nicotine, a main addictive compound in tobacco smoke, has been linked to promotion and progression of lung, head and neck, pancreatic, and breast cancers, but the detailed mechanisms of cancer progression remain elusive. Here, we show that nicotine induces the expression of galectin-3 (an anti-apoptotic β-galactoside-binding lectin) in breast cancer cell line and in primary tumors from breast cancer patients. Nicotine-induced up regulation of galectin-3 is due to an increased expression of α9 isoform of nicotinic acetylcholine receptor (α9nAChR), which activates transcription factor STAT3 that in turn, physically binds to galectin-3 (LGALS3) promoter and induces transcription of galectin-3. Intracellular galectin-3 increased mitochondrial integrity and suppressed chemotherapeutic-induced apoptosis of breast cancer cell. Moreover, nicotine-induced enrichment of side population cells with cancer stem cell-like properties was modulated by galectin-3 expression and could be significantly reduced by transient knock down of LGALS3 and its upstream signaling molecules STAT3 and α9nAChR. Thus, galectin-3 or its upstream signaling molecule STAT3 or α9nAChR could be a potential target to prevent nicotine-induced chemoresistance in breast cancer.
Collapse
Affiliation(s)
- Prasun Guha
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|
6
|
Kalakonda S, Nallar SC, Lindner DJ, Sun P, Lorenz RR, Lamarre E, Reddy SP, Kalvakolanu DV. GRIM-19 mutations fail to inhibit v-Src-induced oncogenesis. Oncogene 2013; 33:3195-204. [PMID: 23851499 PMCID: PMC3916943 DOI: 10.1038/onc.2013.271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 04/25/2013] [Accepted: 04/29/2013] [Indexed: 12/13/2022]
Abstract
The non-receptor tyrosine kinase Src is a major player in multiple physiological responses including growth, survival and differentiation. Overexpression and/or oncogenic mutation in the Src gene have been documented in human tumors. The v-Src protein is an oncogenic mutant of Src, which promotes cell survival, migration, invasion and division. GRIM-19 is an anti-oncogene isolated using a genome-wide knockdown screen. GRIM-19 binds to transcription factor STAT3 and ablates its pro-oncogenic effects while v-Src activates STAT3 to promote its oncogenic effects. However, we found that GRIM-19 inhibits the pro-oncogenic effects of v-Src independently of STAT3. Here, we report the identification of functionally inactivating GRIM-19 mutations in a set of Head and Neck cancer patients. While wild-type GRIM-19 strongly ablated v-Src-induced cell migration, cytoskeletal remodeling and tumor metastasis, the tumor-derived mutants (L71P, L91P and A95T) did not. These mutants were also incapable of inhibiting the drug resistance of v-Src-transformed cells. v-Src down regulated the expression of Pag1, a lipid raft-associated inhibitor of Src, which was restored by wild-type GRIM-19. The tumor-derived mutant GRIM-19 proteins failed to upregulate Pag1. These studies show a novel mechanism that deregulates Src activity in cancer cells.
Collapse
Affiliation(s)
- S Kalakonda
- Department of Microbiology & Immunology, Program in Oncology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - S C Nallar
- Department of Microbiology & Immunology, Program in Oncology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - P Sun
- Department of Microbiology & Immunology, Program in Oncology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - R R Lorenz
- Head & Neck Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - E Lamarre
- Head & Neck Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - S P Reddy
- Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - D V Kalvakolanu
- Department of Microbiology & Immunology, Program in Oncology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
7
|
Ischemia-induced inhibition of mitochondrial complex I in rat brain: effect of permeabilization method and electron acceptor. Neurochem Res 2012; 37:965-76. [PMID: 22219133 DOI: 10.1007/s11064-011-0689-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 12/18/2011] [Accepted: 12/23/2011] [Indexed: 12/23/2022]
Abstract
In this study we have examined the effect of global brain ischemia/reperfusion on biochemical properties of the mitochondrial respiratory complex I (CI) in rat hippocampus and cortex. Since the inner mitochondrial membrane forms the permeability barrier for NADH, the methodology of enzymatic activity determinations employs membrane permeabilization methods. This action affects the basic character of electrostatic and hydrophobic interactions inside the membrane and might influence functional properties of membrane embedded proteins. Therefore we have performed the comparative analysis of two permeabilization methods (sonication, detergent) and their impact on CI enzymatic activities under global brain ischemic-reperfusion conditions. We have observed that ischemia led to significant decrease of CI activities using both permeabilization methods in both brain areas. However, significant differencies in enzymatic activities were registered during reperfusion intervals according to used permeabilization method. We have also tested the effect of electron acceptors (decylubiquinone, potassium ferricyanide, nitrotetrazolium blue) on CI activities during I/R. Based on our results we assume that the critical site where ischemia affects CI activities is electron transfer to electron acceptor. Further, the observed mitochondrial dysfunction was analyzed by means of one and 2-dimensional BN PAGE/SDS PAGE with the focus on 3-nitrotyrosine immunodetection as a marker of oxidative damage to proteins. Add to this, initialization of p53 mitochondrial apoptosis through p53, Bax, Bcl-X(L) proteins and a possible involvement of GRIM-19, the CI structural subunit, in apoptotic processes were also studied.
Collapse
|
8
|
Hao H, Liu J, Liu G, Guan D, Yang Y, Zhang X, Cao X, Liu Q. Depletion of GRIM-19 accelerates hepatocellular carcinoma invasion via inducing EMT and loss of contact inhibition. J Cell Physiol 2011; 227:1212-9. [DOI: 10.1002/jcp.24025] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
9
|
GRIM-1, a novel growth suppressor, inhibits rRNA maturation by suppressing small nucleolar RNAs. PLoS One 2011; 6:e24082. [PMID: 21931644 PMCID: PMC3169572 DOI: 10.1371/journal.pone.0024082] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 07/29/2011] [Indexed: 01/13/2023] Open
Abstract
We have recently isolated novel IFN-inducible gene, Gene associated with Retinoid-Interferon-induced Mortality-1 (GRIM-1), using a genetic technique. Moderate ectopic expression of GRIM-1 caused growth inhibition and sensitized cells to retinoic acid (RA)/IFN-induced cell death while high expression caused apoptosis. GRIM-1 depletion, using RNAi, conferred a growth advantage. Three protein isoforms (1α, 1β and 1γ) with identical C-termini are produced from GRIM-1 mRNA. We show that GRIM-1 isoforms interact with NAF1 and DKC1, two essential proteins required for box H/ACA sno/sca RNP biogenesis and suppresses box H/ACA RNA levels in mammalian cells by delocalizing NAF1. Suppression of these small RNAs manifests as inefficient rRNA maturation and growth suppression. Interestingly, yeast Shq1p also caused growth suppression in mammalian cells. Consistent with its growth-suppressive property, GRIM-1 expression is lost in a number of human primary prostate tumors. Our observations support a recent study that GRIM-1 might act as a co-tumor suppressor in the prostate.
Collapse
|
10
|
Moreira S, Correia M, Soares P, Máximo V. GRIM-19 function in cancer development. Mitochondrion 2011; 11:693-9. [DOI: 10.1016/j.mito.2011.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 04/05/2011] [Accepted: 05/25/2011] [Indexed: 02/04/2023]
|
11
|
GRIM-19 disrupts E6/E6AP complex to rescue p53 and induce apoptosis in cervical cancers. PLoS One 2011; 6:e22065. [PMID: 21765936 PMCID: PMC3134474 DOI: 10.1371/journal.pone.0022065] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Accepted: 06/14/2011] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Our previous studies showed a down-regulation of GRIM-19 in primary human cervical cancers, and restoration of GRIM-19 induced tumor regression. The induction of tumor suppressor protein p53 ubiquitination and degradation by E6 oncoportein of high risk-HPV through forming a stable complex with E6AP is considered as a critical mechanism for cervical tumor development. The aims of this study were to determine the potential role of GRIM-19 in rescuing p53 protein and inducing cervical cancer cell apoptosis. METHODOLOGY/PRINCIPAL FINDINGS The protein levels of GRIM-19 and p53 were detected in normal cervical tissues from 45 patients who underwent hysterectomy for reasons other than neoplasias of either the cervix or endometrium, and cervical cancer tissues from 60 patients with non-metastatic squamous epithelial carcinomas. Coimmunoprecipitation and GST pull-down assay were performed to examine the interaction of GRIM-19 with 18E6 and E6AP in vivo and in vitro respectively. The competition of 18E6 with E6AP in binding GRIM-19 by performing competition pull-down assays was designed to examine the disruption of E6/E6AP complex by GRIM-19. The augment of E6AP ubiquitination by GRIM-19 was detected in vivo and in vitro ubiquitination assay. The effects of GRIM-19-dependent p53 accumulation on cell proliferation, cell cycle, apoptosis were explored by MTT, flow cytometry and transmission electron microscopy respectively. The tumor suppression was detected by xenograft mouse model. CONCLUSION/SIGNIFICANCE The levels of GRIM-19 and p53 were concurrently down regulated in cervical cancers. The restoration of GRIM-19 can induce ubiquitination and degradation of E6AP, and disrupt the E6/E6AP complex through the interaction of N-terminus of GRIM-19 with both E6 and E6AP, which protected p53 from degradation and promoted cell apoptosis. Tumor xenograft studies also revealed the suppression of p53 degradation in presence of GRIM-19. These data suggest that GRIM-19 can block E6/E6AP complex; and synergistically suppress cervical tumor growth with p53.
Collapse
|
12
|
Sun P, Nallar SC, Raha A, Kalakonda S, Velalar CN, Reddy SP, Kalvakolanu DV. GRIM-19 and p16(INK4a) synergistically regulate cell cycle progression and E2F1-responsive gene expression. J Biol Chem 2010; 285:27545-52. [PMID: 20522552 DOI: 10.1074/jbc.m110.105767] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
GRIM-19 (Gene associated with Retinoid-IFN-induced Mortality-19) was originally isolated as a growth suppressor in a genome-wide knockdown screen with antisense libraries. Like classical tumor suppressors, mutations, and/or loss of GRIM-19 expression occur in primary human tumors; and it is inactivated by viral gene products. Our search for potential GRIM-19-binding proteins, using mass spectrometry, that permit its antitumor actions led to the inhibitor of cyclin-dependent kinase 4, CDKN2A. The GRIM-19/CDKN2A synergistically suppressed cell cycle progression via inhibiting E2F1-driven gene expression. The N terminus of GRIM-19 and the fourth ankyrin repeat of CDKN2A are crucial for their interaction. The biological relevance of these interactions is underscored by observations that GRIM-19 promotes the inhibitory effect of CDKN2A on CDK4; and mutations from primary tumors disrupt its ability to interact with GRIM-19 and suppress E2F1-driven gene expression.
Collapse
Affiliation(s)
- Peng Sun
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Identification of alternatively spliced GRIM-19 mRNA in kidney cancer tissues. J Hum Genet 2010; 55:507-11. [DOI: 10.1038/jhg.2010.57] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
14
|
Kalvakolanu DV, Nallar SC, Kalakonda S. Cytokine-induced tumor suppressors: a GRIM story. Cytokine 2010; 52:128-42. [PMID: 20382543 DOI: 10.1016/j.cyto.2010.03.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Accepted: 03/16/2010] [Indexed: 12/18/2022]
Abstract
Cytokines belonging to the IFN family are potent growth suppressors. In a number of clinical and preclinical studies, vitamin A and its derivatives like retinoic acid (RA) have been shown to exert synergistic growth-suppressive effects on several tumor cells. We have employed a genome-wide expression-knockout approach to identify the genes critical for IFN/RA-induced growth suppression. A number of novel genes associated with Retinoid-Interferon-induced Mortality (GRIM) were isolated. In this review, we will describe the molecular mechanisms of actions of one, GRIM-19, which participates in multiple pathways for exerting growth control and/or cell death. This protein is emerging as a new tumor suppressor. In addition, GRIM-19 appears to participate in innate immune responses as its activity is modulated by several viruses and bacteria. Thus, GRIMs seem to couple with multiple biological responses by acting at critical nodes.
Collapse
Affiliation(s)
- Dhan V Kalvakolanu
- Department of Microbiology & Immunology, Marlene & Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | |
Collapse
|
15
|
Potula HSK, Wang D, Quyen DV, Singh NK, Kundumani-Sridharan V, Karpurapu M, Park EA, Glasgow WC, Rao GN. Src-dependent STAT-3-mediated expression of monocyte chemoattractant protein-1 is required for 15(S)-hydroxyeicosatetraenoic acid-induced vascular smooth muscle cell migration. J Biol Chem 2009; 284:31142-55. [PMID: 19736311 DOI: 10.1074/jbc.m109.012526] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
To understand the role of human 15-lipoxygenase 1 (15-LOX1) in vascular wall remodeling, we have studied the effect of the major 15-LOX1 metabolite of arachidonic acid, 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE), on vascular smooth muscle cell (VSMC) migration both in vitro and in vivo. Among 5(S)-HETE, 12(S)-HETE, and 15(S)-HETE, 15(S)-HETE potentially stimulated more vascular smooth muscle cell (VSMC) migration. In addition, 15(S)-HETE-induced VSMC migration was dependent on Src-mediated activation of signal transducer and activator of transcription-3 (STAT-3). 15(S)-HETE also induced monocyte chemoattractant protein-1 (MCP-1) expression via Src-STAT-3 signaling, and neutralizing anti-MCP-1 antibodies completely negated 15(S)-HETE-induced VSMC migration. Cloning and characterization of a 2.6-kb MCP-1 promoter revealed the presence of four putative STAT-binding sites, and the site that is proximal to the transcription start site was found to be essential for 15(S)-HETE-induced Src-STAT-3-mediated MCP-1 expression. Rat carotid arteries that were subjected to balloon injury and transduced with Ad-15-LOX1 upon exposure to [(3)H]arachidonic acid ex vivo produced 15-HETE as a major eicosanoid and enhanced balloon injury-induced expression of MCP-1 in smooth muscle cells in Src and STAT-3-dependent manner in vivo. Adenovirus-mediated delivery of 15-LOX1 into rat carotid artery also led to recruitment and homing of macrophages to medial region in response to injury. In addition, transduction of Ad-15-LOX1 into arteries enhanced balloon injury-induced smooth muscle cell migration from media to intima and neointima formation. These results show for the first time that 15-LOX1-15(S)-HETE axis plays a major role in vascular wall remodeling after balloon angioplasty.
Collapse
Affiliation(s)
- Harihara S K Potula
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Galluzzi L, Morselli E, Kepp O, Vitale I, Rigoni A, Vacchelli E, Michaud M, Zischka H, Castedo M, Kroemer G. Mitochondrial gateways to cancer. Mol Aspects Med 2009; 31:1-20. [PMID: 19698742 DOI: 10.1016/j.mam.2009.08.002] [Citation(s) in RCA: 190] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Revised: 08/12/2009] [Accepted: 08/13/2009] [Indexed: 12/29/2022]
Abstract
Mitochondria are required for cellular survival, yet can also orchestrate cell death. The peculiar biochemical properties of these organelles, which are intimately linked to their compartmentalized ultrastructure, provide an optimal microenvironment for multiple biosynthetic and bioenergetic pathways. Most intracellular ATP is generated by mitochondrial respiration, which also represents the most relevant source of intracellular reactive oxygen species. Mitochondria participate in a plethora of anabolic pathways, including cholesterol, cardiolipin, heme and nucleotide biosynthesis. Moreover, mitochondria integrate numerous pro-survival and pro-death signals, thereby exerting a decisive control over several biochemical cascades leading to cell death, in particular the intrinsic pathway of apoptosis. Therefore, it is not surprising that cancer cells often manifest the deregulation of one or several mitochondrial functions. The six classical hallmarks of cancer (i.e., limitless replication, self-provision of proliferative stimuli, insensitivity to antiproliferative signals, disabled apoptosis, sustained angiogenesis, invasiveness/metastatic potential), as well as other common features of tumors (i.e., avoidance of the immune response, enhanced anabolic metabolism, disabled autophagy) may directly or indirectly implicate deregulated mitochondria. In this review, we discuss several mechanisms by which mitochondria can contribute to malignant transformation and tumor progression.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- INSERM, U848, Institut Gustave Roussy, PR1, 39 Rue Camille Desmoulins, F-94805 Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|