1
|
Xie G, Li Y, Jiang Y, Ye X, Tang J, Chen J. Silencing HIPPI Suppresses Tumor Progression in Non-Small-Cell Lung Cancer by Inhibiting DNA Replication. Onco Targets Ther 2021; 14:3467-3480. [PMID: 34079292 PMCID: PMC8166357 DOI: 10.2147/ott.s305388] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/28/2021] [Indexed: 12/09/2022] Open
Abstract
Introduction Non-small cell lung cancer (NSCLC) is the most common form of lung cancer, accounting for approximately 80%-85% of all cases of lung cancer. Huntingtin interacting protein-1 interacting protein (HIPPI) is a transcription regulator and plays an important role in apoptotic cell death. However, the role of HIPPI in NSCLC remains unclear. Methods Immunohistochemistry (IHC) and qRT-PCR were performed for expression analysis. The roles of HIPPI were studied using cell counting kit-8 (CCK-8), colony formation, flow cytometry, wound healing, Transwell invasion assays and mouse xenograft model. Gene microarray analysis and bioinformatics analysis were used to identify differentially expressed genes after HIPPI silencing. Results HIPPI is highly expressed in NSCLC tissues relative to adjacent normal tissues. Targeting HIPPI by RNA interference inhibits NSCLC cell proliferation in vitro and tumor growth in vivo. HIPPI silencing also attenuates cell migration and invasion and enhances cisplatin sensitivity in NSCLC cells. Mechanistic investigation suggests that HIPPI can positively regulate the expression of MCM2, MCM6 and MCM8, which are key regulators of DNA replication. Furthermore, consistent with HIPPI, MCM2, MCM6 and MCM8 are also upregulated in NSCLC tissues. Conclusion Our study highlights the importance of HIPPI for tumor biology in NSCLC and suggests that HIPPI may be a potential therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Guanghui Xie
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Department of Cardiothoracic Vascular Surgery, The Central Hospital of Yongzhou, Yongzhou, Hunan Province, People's Republic of China
| | - Yongwen Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yongjun Jiang
- Department of Cardiothoracic Vascular Surgery, The Central Hospital of Yongzhou, Yongzhou, Hunan Province, People's Republic of China
| | - Xian Ye
- Department of Cardiothoracic Vascular Surgery, The Central Hospital of Yongzhou, Yongzhou, Hunan Province, People's Republic of China
| | - Jianfeng Tang
- Department of Cardiothoracic Vascular Surgery, The Central Hospital of Yongzhou, Yongzhou, Hunan Province, People's Republic of China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| |
Collapse
|
2
|
Zhang Y, Zhang X, Cheng A, Wang M, Yin Z, Huang J, Jia R. Apoptosis Triggered by ORF3 Proteins of the Circoviridae Family. Front Cell Infect Microbiol 2021; 10:609071. [PMID: 33604306 PMCID: PMC7884757 DOI: 10.3389/fcimb.2020.609071] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Apoptosis, a form of the programmed cell death, is an indispensable defense mechanism regulating cellular homeostasis and is triggered by multiple stimuli. Because of the regulation of apoptosis in cellular homeostasis, viral proteins with apoptotic activity are particular foci of on antitumor therapy. One representative viral protein is the open reading frame 3 (ORF3) protein, also named as apoptin in the Circoviridae chicken anemia virus (CAV), and has the ability to induce tumor-specific apoptosis. Proteins encoded by ORF3 in other circovirus species, such as porcine circovirus (PCV) and duck circovirus (DuCV), have also been reported to induce apoptosis, with subtle differences in apoptotic activity based on cell types. This article is aimed at reviewing the latest research advancements in understanding ORF3 protein-mediated apoptosis mechanisms of Circoviridae from three perspectives: subcellular localization, interactions with host proteins, and participation in multiple apoptotic signaling pathways, providing a scientific basis for circovirus pathogenesis and a reference on its potential anticancer function.
Collapse
Affiliation(s)
- Yanting Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xingcui Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhongqiong Yin
- Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Juan Huang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
3
|
Tiys ES, Ivanisenko TV, Demenkov PS, Ivanisenko VA. FunGeneNet: a web tool to estimate enrichment of functional interactions in experimental gene sets. BMC Genomics 2018; 19:76. [PMID: 29504895 PMCID: PMC5836822 DOI: 10.1186/s12864-018-4474-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Estimation of functional connectivity in gene sets derived from genome-wide or other biological experiments is one of the essential tasks of bioinformatics. A promising approach for solving this problem is to compare gene networks built using experimental gene sets with random networks. One of the resources that make such an analysis possible is CrossTalkZ, which uses the FunCoup database. However, existing methods, including CrossTalkZ, do not take into account individual types of interactions, such as protein/protein interactions, expression regulation, transport regulation, catalytic reactions, etc., but rather work with generalized types characterizing the existence of any connection between network members. Results We developed the online tool FunGeneNet, which utilizes the ANDSystem and STRING to reconstruct gene networks using experimental gene sets and to estimate their difference from random networks. To compare the reconstructed networks with random ones, the node permutation algorithm implemented in CrossTalkZ was taken as a basis. To study the FunGeneNet applicability, the functional connectivity analysis of networks constructed for gene sets involved in the Gene Ontology biological processes was conducted. We showed that the method sensitivity exceeds 0.8 at a specificity of 0.95. We found that the significance level of the difference between gene networks of biological processes and random networks is determined by the type of connections considered between objects. At the same time, the highest reliability is achieved for the generalized form of connections that takes into account all the individual types of connections. By taking examples of the thyroid cancer networks and the apoptosis network, it is demonstrated that key participants in these processes are involved in the interactions of those types by which these networks differ from random ones. Conclusions FunGeneNet is a web tool aimed at proving the functionality of networks in a wide range of sizes of experimental gene sets, both for different global networks and for different types of interactions. Using examples of thyroid cancer and apoptosis networks, we have shown that the links over-represented in the analyzed network in comparison with the random ones make possible a biological interpretation of the original gene/protein sets. The FunGeneNet web tool for assessment of the functional enrichment of networks is available at http://www-bionet.sscc.ru/fungenenet/. Electronic supplementary material The online version of this article (10.1186/s12864-018-4474-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Evgeny S Tiys
- The Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Prospekt Lavrentyeva 10, 630090, Novosibirsk, Russia. .,Laboratory of Computer Genomics, Novosibirsk State University, Pirogova Str. 2, 630090, Novosibirsk, Russia.
| | - Timofey V Ivanisenko
- The Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Prospekt Lavrentyeva 10, 630090, Novosibirsk, Russia.,Laboratory of Computer Genomics, Novosibirsk State University, Pirogova Str. 2, 630090, Novosibirsk, Russia
| | - Pavel S Demenkov
- The Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Prospekt Lavrentyeva 10, 630090, Novosibirsk, Russia
| | - Vladimir A Ivanisenko
- The Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Prospekt Lavrentyeva 10, 630090, Novosibirsk, Russia
| |
Collapse
|
4
|
Banerjee M, Datta M, Bhattacharyya NP. Modulation of mutant Huntingtin aggregates and toxicity by human myeloid leukemia factors. Int J Biochem Cell Biol 2016; 82:1-9. [PMID: 27840155 DOI: 10.1016/j.biocel.2016.11.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 10/26/2016] [Accepted: 11/09/2016] [Indexed: 12/28/2022]
Abstract
Increased poly glutamine (polyQ) stretch at N-terminal of Huntingtin (HTT) causes Huntington's disease. HTT interacts with large number of proteins, although the preference for such interactions with wild type or mutated HTT protein remains largely unknown. HYPK, an intrinsically unstructured protein chaperone and interactor of mutant HTT was found to interact with myeloid leukemia factor 1 (MLF1) and 2 (MLF2). To identify the role of these two proteins in mutant HTT mediated aggregate formation and toxicity in a cell model, both the proteins were found to preferentially interact with the mutated N-terminal HTT. They significantly reduced the number of cells containing mutant HTT aggregates and subsequent apoptosis in Neuro2A cells. Additionally, in FRAP assay, mobile fraction of mutant HTT aggregates was increased in the presence of MLF1 or MLF2. Further, MLF1 could release transcription factors like p53, CBP and CREB from mutant HTT aggregates. Moreover, in HeLa cell co-expressing mutant HTT exon1 and full length MLF1, p53 was released from the aggregates, leading to the recovery of the expression of the GADD45A transcript, a p53 regulated gene. Taking together, these results showed that MLF1 and MLF2 modulated the formation of aggregates and induction of apoptosis as well as the expressions of genes indirectly.
Collapse
Affiliation(s)
- Manisha Banerjee
- Crystallography & Molecular Biology Division and Structural Genomics Section, Saha Institute of Nuclear Physics, 1/AF, Bidhan Nagar, Kolkata, 700064, India.
| | - Moumita Datta
- Crystallography & Molecular Biology Division and Structural Genomics Section, Saha Institute of Nuclear Physics, 1/AF, Bidhan Nagar, Kolkata, 700064, India
| | - Nitai P Bhattacharyya
- Crystallography & Molecular Biology Division and Structural Genomics Section, Saha Institute of Nuclear Physics, 1/AF, Bidhan Nagar, Kolkata, 700064, India.
| |
Collapse
|
5
|
Exploring Genetic Factors Involved in Huntington Disease Age of Onset: E2F2 as a New Potential Modifier Gene. PLoS One 2015; 10:e0131573. [PMID: 26148071 PMCID: PMC4493078 DOI: 10.1371/journal.pone.0131573] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 06/03/2015] [Indexed: 01/09/2023] Open
Abstract
Age of onset (AO) of Huntington disease (HD) is mainly determined by the length of the CAG repeat expansion (CAGexp) in exon 1 of the HTT gene. Additional genetic variation has been suggested to contribute to AO, although the mechanism by which it could affect AO is presently unknown. The aim of this study is to explore the contribution of candidate genetic factors to HD AO in order to gain insight into the pathogenic mechanisms underlying this disorder. For that purpose, two AO definitions were used: the earliest age with unequivocal signs of HD (earliest AO or eAO), and the first motor symptoms age (motor AO or mAO). Multiple linear regression analyses were performed between genetic variation within 20 candidate genes and eAO or mAO, using DNA and clinical information of 253 HD patients from REGISTRY project. Gene expression analyses were carried out by RT-qPCR with an independent sample of 35 HD patients from Basque Country Hospitals. We found suggestive association signals between HD eAO and/or mAO and genetic variation within the E2F2, ATF7IP, GRIN2A, GRIN2B, LINC01559, HIP1 and GRIK2 genes. Among them, the most significant was the association between eAO and rs2742976, mapping to the promoter region of E2F2 transcription factor. Furthermore, rs2742976 T allele patient carriers exhibited significantly lower lymphocyte E2F2 gene expression, suggesting a possible implication of E2F2-dependent transcriptional activity in HD pathogenesis. Thus, E2F2 emerges as a new potential HD AO modifier factor.
Collapse
|
6
|
Raychaudhuri S, Banerjee R, Mukhopadhyay S, Bhattacharyya NP. Conserved C-terminal nascent peptide binding domain of HYPK facilitates its chaperone-like activity. J Biosci 2015; 39:659-72. [PMID: 25116620 DOI: 10.1007/s12038-014-9442-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Human HYPK (Huntingtin Yeast-two-hybrid Protein K) is an intrinsically unstructured chaperone-like protein with no sequence homology to known chaperones. HYPK is also known to be a part of ribosome-associated protein complex and present in polysomes. The objective of the present study was to investigate the evolutionary influence on HYPK primary structure and its impact on the protein's function. Amino acid sequence analysis revealed 105 orthologs of human HYPK from plants, lower invertebrates to mammals. C-terminal part of HYPK was found to be particularly conserved and to contain nascent polypeptide-associated alpha subunit (NPAA) domain. This region experiences highest selection pressure, signifying its importance in the structural and functional evolution. NPAA domain of human HYPK has unique amino acid composition preferring glutamic acid and happens to be more stable from a conformational point of view having higher content of a-helices than the rest. Cell biology studies indicate that overexpressed C-terminal human HYPK can interact with nascent proteins, co-localizes with huntingtin, increases cell viability and decreases caspase activities in Huntington's disease (HD) cell culture model. This domain is found to be required for the chaperone-like activity of HYPK in vivo. Our study suggested that by virtue of its flexibility and nascent peptide binding activity, HYPK may play an important role in assisting protein (re)folding.
Collapse
Affiliation(s)
- Swasti Raychaudhuri
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata 700 064, India,
| | | | | | | |
Collapse
|
7
|
Schlachetzki JC, Saliba SW, Oliveira ACPD. Studying neurodegenerative diseases in culture models. BRAZILIAN JOURNAL OF PSYCHIATRY 2013; 35 Suppl 2:S92-100. [DOI: 10.1590/1516-4446-2013-1159] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
8
|
Hyman BT, Yuan J. Apoptotic and non-apoptotic roles of caspases in neuronal physiology and pathophysiology. Nat Rev Neurosci 2012; 13:395-406. [PMID: 22595785 DOI: 10.1038/nrn3228] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Caspases are cysteine proteases that mediate apoptosis, which is a form of regulated cell death that effectively and efficiently removes extra and unnecessary cells during development. In the mature nervous system, caspases are not only involved in mediating cell death but also regulatory events that are important for neural functions, such as axon pruning and synapse elimination, which are necessary to refine mature neuronal circuits. Furthermore, caspases can be reactivated to cause cell death as well as non-lethal changes in neurons during numerous pathological processes. Thus, although a global activation of caspases leads to apoptosis, restricted and localized activation may control normal physiology and pathophysiology in living neurons. This Review explores the multiple roles of caspase activity in neurons.
Collapse
Affiliation(s)
- Bradley T Hyman
- Neurology Service, Massachusetts General Hospital, 114 16th Street Charlestown, Massachusetts 01029, USA.
| | | |
Collapse
|
9
|
Datta M, Choudhury A, Lahiri A, Bhattacharyya NP. Genome wide gene expression regulation by HIP1 Protein Interactor, HIPPI: prediction and validation. BMC Genomics 2011; 12:463. [PMID: 21943362 PMCID: PMC3228557 DOI: 10.1186/1471-2164-12-463] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 09/26/2011] [Indexed: 12/04/2022] Open
Abstract
Background HIP1 Protein Interactor (HIPPI) is a pro-apoptotic protein that induces Caspase8 mediated apoptosis in cell. We have shown earlier that HIPPI could interact with a specific 9 bp sequence motif, defined as the HIPPI binding site (HBS), present in the upstream promoter of Caspase1 gene and regulate its expression. We also have shown that HIPPI, without any known nuclear localization signal, could be transported to the nucleus by HIP1, a NLS containing nucleo-cytoplasmic shuttling protein. Thus our present work aims at the investigation of the role of HIPPI as a global transcription regulator. Results We carried out genome wide search for the presence of HBS in the upstream sequences of genes. Our result suggests that HBS was predominantly located within 2 Kb upstream from transcription start site. Transcription factors like CREBP1, TBP, OCT1, EVI1 and P53 half site were significantly enriched in the 100 bp vicinity of HBS indicating that they might co-operate with HIPPI for transcription regulation. To illustrate the role of HIPPI on transcriptome, we performed gene expression profiling by microarray. Exogenous expression of HIPPI in HeLa cells resulted in up-regulation of 580 genes (p < 0.05) while 457 genes were down-regulated. Several transcription factors including CBP, REST, C/EBP beta were altered by HIPPI in this study. HIPPI also interacted with P53 in the protein level. This interaction occurred exclusively in the nuclear compartment and was absent in cells where HIP1 was knocked down. HIPPI-P53 interaction was necessary for HIPPI mediated up-regulation of Caspase1 gene. Finally, we analyzed published microarray data obtained with post mortem brains of Huntington's disease (HD) patients to investigate the possible involvement of HIPPI in HD pathogenesis. We observed that along with the transcription factors like CREB, P300, SREBP1, Sp1 etc. which are already known to be involved in HD, HIPPI binding site was also significantly over-represented in the upstream sequences of genes altered in HD. Conclusions Taken together, the results suggest that HIPPI could act as an important transcription regulator in cell regulating a vast array of genes, particularly transcription factors and at least, in part, play a role in transcription deregulation observed in HD.
Collapse
Affiliation(s)
- Moumita Datta
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, 1/AF Bidhan Nagar, Kolkata 700 064, India
| | | | | | | |
Collapse
|
10
|
Datta M, Bhattacharyya NP. Regulation of RE1 protein silencing transcription factor (REST) expression by HIP1 protein interactor (HIPPI). J Biol Chem 2011; 286:33759-69. [PMID: 21832040 DOI: 10.1074/jbc.m111.265173] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Earlier we have shown that the proapoptotic protein HIPPI (huntingtin interacting protein 1 (HIP1) protein interactor) along with its molecular partner HIP1 could regulate transcription of the caspase-1 gene. Here we report that RE1-silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) is a new transcriptional target of HIPPI. HIPPI could bind to the promoter of REST and increased its expression in neuronal as well as non-neuronal cells. Such activation of REST down-regulated expression of REST target genes, such as brain-derived neurotrophic factor (BDNF) or proenkephalin (PENK). The ability of HIPPI to activate REST gene transcription was dependent on HIP1, the nuclear transporter of HIPPI. Using a Huntington disease cell model, we have demonstrated that feeble interaction of HIP1 with mutant huntingtin protein resulted in increased nuclear accumulation of HIPPI and HIP1, leading to higher occupancy of HIPPI at the REST promoter, triggering its transcriptional activation and consequent repression of REST target genes. This novel transcription regulatory mechanism of REST by HIPPI may contribute to the deregulation of transcription observed in the cell model of Huntington disease.
Collapse
Affiliation(s)
- Moumita Datta
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, 1/AF Bidhan Nagar, Kolkata 700 064, India
| | | |
Collapse
|
11
|
Banerjee M, Datta M, Majumder P, Mukhopadhyay D, Bhattacharyya NP. Transcription regulation of caspase-1 by R393 of HIPPI and its molecular partner HIP-1. Nucleic Acids Res 2009; 38:878-92. [PMID: 19934260 PMCID: PMC2817453 DOI: 10.1093/nar/gkp1011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Earlier we have shown that exogenous expression of HIPPI, a molecular partner of Huntingtin interacting protein HIP-1, induces apoptosis and increases expression of caspases-1, -8 and -10 in HeLa and Neuro2A cells. The C-terminal pseudo death effector domain of HIPPI (pDED-HIPPI) specifically interacts with the putative promoter sequences of these genes. In the present manuscript, we predict from structural modeling of pDED-HIPPI that R393 of HIPPI is important for such interaction. R393E mutation in pDED-HIPPI decreases the interaction with the putative promoter of caspase-1 in cells. Expression of caspase-1 is decreased in cells expressing mutant pDED-HIPPI in comparison to that observed in cells expressing wild type pDED-HIPPI. Using HIP-1 knocked down cells as well as over expressing HIP-1 with mutation at its nuclear localization signal and other deletion mutations, we demonstrate that translocation of HIPPI to the nucleus is mediated by HIP-1 for the increased expression of caspase-1. HIPPI-HIP-1 heterodimer is detected in cytoplasm as well as in the nucleus and is associated with transcription complex in cells. Taking together, we are able to show the importance of R393 of HIPPI and the role of HIPPI-HIP-1 heterodimer in the transcription regulation of caspase-1.
Collapse
Affiliation(s)
- M Banerjee
- Saha Institute of Nuclear Physics, 1/AF Bidhan Nagar, Kolkata 700 064, India
| | | | | | | | | |
Collapse
|
12
|
Browne SE. Mitochondria and Huntington's disease pathogenesis: insight from genetic and chemical models. Ann N Y Acad Sci 2009; 1147:358-82. [PMID: 19076457 DOI: 10.1196/annals.1427.018] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
A mechanistic link between cellular energetic defects and the pathogenesis of Huntington's disease (HD) has long been hypothesized based on the cardinal observations of progressive weight loss in patients and metabolic defects in brain and muscle. Identification of respiratory chain deficits in HD postmortem brain led to the use of mitochondrial complex II inhibitors to generate acute toxicity models that replicate aspects of HD striatal pathology in vivo. Subsequently, the generation of progressive genetic animal models has enabled characterization of numerous cellular and systematic changes over disease etiology, including mitochondrial modifications that impact cerebral metabolism, calcium handling, oxidative damage, and apoptotic cascades. This review focuses on how HD animal models have influenced our understanding of mechanisms underlying HD pathogenesis, concentrating on insight gained into the roles of mitochondria in disease etiology. One outstanding question concerns the hierarchy of mitochondrial alterations in the cascade of events following mutant huntingtin (mhtt)-induced toxicity. One hypothesis is that a direct interaction of mhtt with mitochondria may trigger the neuronal damage and degeneration that occurs in HD. While there is evidence that mhtt associates with mitochondria, deleterious consequences of this interaction have not yet been established. Contrary evidence suggests that a primary nuclear action of mhtt may detrimentally influence mitochondrial function via effects on gene transcription. Irrespective of whether the principal toxic action of mhtt directly or secondarily impacts mitochondria, the repercussions of sufficient mitochondrial dysfunction are catastrophic to cells and may arguably underlie many of the other disruptions in cellular processes that evolve during HD pathogenesis.
Collapse
|
13
|
Gdynia G, Lehmann-Koch J, Sieber S, Tagscherer KE, Fassl A, Zentgraf H, Matsuzawa SI, Reed JC, Roth W. BLOC1S2 interacts with the HIPPI protein and sensitizes NCH89 glioblastoma cells to apoptosis. Apoptosis 2008; 13:437-47. [PMID: 18188704 DOI: 10.1007/s10495-007-0176-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The HIPPI (HIP-1 protein interactor) protein is a multifunctional protein that is involved in the regulation of apoptosis. The interaction partners of HIPPI include HIP-1 (Huntingtin-interacting protein-1), Apoptin, Homer1c, Rybp/DEDAF, and BAR (bifunctional apoptosis regulator). In search for other binding partners of HIPPI, we performed a yeast two hybrid screen and identified BLOC1S2 (Biogenesis of lysosome-related organelles complex-1 subunit 2) as a novel HIPPI-interacting protein. In co-immunoprecipitation assays, BLOC1S2 specifically associates with HIPPI, but not with HIP-1. To study the expression of BLOC1S2 on the protein level, we generated a mouse monoclonal antibody specific for BLOC1S2 and a multiple tissue array comprising 70 normal and cancer tissue samples of diverse origin. BLOC1S2 protein is widely expressed in normal tissue as well as in malignant tumors with a tendency towards lower expression levels in certain subtypes of tumors. On the subcellular level, BLOC1S2 is expressed in an organellar-like pattern and co-localizes with mitochondria. Over-expression of BLOC1S2 in the presence or absence of HIPPI does not induce apoptosis. However, BLOC1S2 and HIPPI sensitize NCH89 glioblastoma cells to the pro-apoptotic actions of staurosporine and the death ligand TRAIL by enhancing caspase activation, cytochrome c release, and disruption of the mitochondrial membrane potential. Given its interaction with HIPPI and its pro-apoptotic activity, BLOC1S2 might play an important functional role in cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Georg Gdynia
- Molecular Neuro-Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Bhattacharyya NP, Banerjee M, Majumder P. Huntington’s disease: roles of huntingtin-interacting protein 1 (HIP-1) and its molecular partner HIPPI in the regulation of apoptosis and transcription. FEBS J 2008; 275:4271-9. [DOI: 10.1111/j.1742-4658.2008.06563.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
15
|
Backendorf C, Visser AE, de Boer AG, Zimmerman R, Visser M, Voskamp P, Zhang YH, Noteborn M. Apoptin: therapeutic potential of an early sensor of carcinogenic transformation. Annu Rev Pharmacol Toxicol 2008; 48:143-69. [PMID: 17848136 DOI: 10.1146/annurev.pharmtox.48.121806.154910] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The avian virus-derived protein apoptin induces p53-independent apoptosis in a tumor-specific way. Apoptin acts as a multimeric complex and forms superstructures upon binding to DNA. In tumor cells, apoptin is phosphorylated and mainly nuclear, whereas in normal cells it is unphosphorylated, cytoplasmic, and becomes readily neutralized. Interestingly, apoptin phosphorylation, nuclear translocation, and apoptosis can transiently be induced in normal cells by cotransfecting SV40 large T oncogene, indicating that apoptin recognizes early stages of oncogenic transformation. In cancer cells, apoptin appears to recognize survival signals, which it is able to redirect into cell death impulses. Apoptin targets include DEDAF, Nur77, Nmi, Hippi, and the potential drug target APC1. Apoptin-transgenic mice and animal tumor models have revealed apoptin as a safe and efficient antitumor agent, resulting in significant tumor regression. Future antitumor therapies could use apoptin either as a therapeutic bullet or as an early sensor of druggable tumor-specific processes.
Collapse
Affiliation(s)
- Claude Backendorf
- Molecular Genetics, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Majumder P, Raychaudhuri S, Chattopadhyay B, Bhattacharyya NP. Increased caspase-2, calpain activations and decreased mitochondrial complex II activity in cells expressing exogenous huntingtin exon 1 containing CAG repeat in the pathogenic range. Cell Mol Neurobiol 2007; 27:1127-45. [PMID: 17902043 DOI: 10.1007/s10571-007-9220-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Accepted: 08/31/2007] [Indexed: 11/28/2022]
Abstract
(1) Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by the expansion of polymorphic CAG repeats beyond 36 at exon 1 of huntingtin gene (htt). To study cellular effects by expressing N-terminal domain of Huntingtin (Htt) in specific cell lines, we expressed exon 1 of htt that codes for 40 glutamines (40Q) and 16Q in Neuro2A and HeLa cells. (2) Aggregates and various apoptotic markers were detected at various time points after transfection. In addition, we checked the alterations of expressions of few apoptotic genes by RT-PCR. (3) Cells expressing exon 1 of htt coding 40Q at a stretch exhibited nuclear and cytoplasmic aggregates, increased caspase-1, caspase-2, caspase-8, caspase-9/6, and calpain activations, release of cytochrome c and AIF from mitochondria in a time-dependent manner. Truncation of Bid was increased, while the activity of mitochondrial complex II was decreased in such cells. These changes were significantly higher in cells expressing N-terminal Htt with 40Q than that obtained in cells expressing N-terminal Htt with 16Q. Expressions of caspase-1, caspase-2, caspase-3, caspase-7, and caspase-8 were increased while expression of Bcl-2 was decreased in cells expressing mutated Htt-exon 1. (4) Results presented in this communication showed that expression of mutated Htt-exon 1 could mimic the cellular phenotypes observed in Huntington's disease and this cell model can be used for screening the agents that would interfere with the apoptotic pathway and aggregate formation.
Collapse
Affiliation(s)
- Pritha Majumder
- Saha Institute of Nuclear Physics, 1/AF Bidhan Nagar, Kolkata, 700064, India
| | | | | | | |
Collapse
|
17
|
HYPK, a Huntingtin interacting protein, reduces aggregates and apoptosis induced by N-terminal Huntingtin with 40 glutamines in Neuro2a cells and exhibits chaperone-like activity. Hum Mol Genet 2007; 17:240-55. [DOI: 10.1093/hmg/ddm301] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
18
|
Stanton SE, Blanck JK, Locker J, Schreiber-Agus N. Rybp interacts with Hippi and enhances Hippi-mediated apoptosis. Apoptosis 2007; 12:2197-206. [PMID: 17874297 DOI: 10.1007/s10495-007-0131-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Rybp (DEDAF) has been shown to interact with DED-containing proteins and to encode pro-apoptotic functions. Here we characterize a novel interaction between Rybp and Hippi, a protein implicated in neuronal apoptosis as well as in the pathogenesis of Huntington's disease. Rybp can synergize with Hippi to enhance Caspase 8-mediated apoptosis and also appears to be essential for Hippi-mediated apoptosis. Moreover, Rybp may mediate or regulate the interaction between Hippi and Caspase 8. Finally, Rybp and Hippi co-localize in a subset of neurons in the developing mouse brain. Together, these findings suggest that Rybp and Hippi may functionally interact in the apoptotic processes that accompany normal murine neural development.
Collapse
Affiliation(s)
- Sasha E Stanton
- Department of Molecular Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Ullmann 809, Bronx, NY 10461, USA.
| | | | | | | |
Collapse
|
19
|
Visser AE, Backendorf C, Noteborn M. Viral protein apoptin as a molecular tool and therapeutic bullet: implications for cancer control. Future Virol 2007. [DOI: 10.2217/17460794.2.5.519] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The chicken anemia virus-derived protein apoptin induces apoptosis in human tumor cells via a p53-independent pathway, while leaving normal cells intact. Moreover, apoptin treatment in preclinical animal studies leads to reduced tumor growth or remission without a detectable effect on healthy tissues. Apoptin is activated by a still unknown tumor-specific kinase activity. The mode of action of apoptin is under intense investigation, as certain features make it a promising tool for discovering early events in tumorigenesis, identifying druggable targets for antitumor treatment and possibly serving as an antitumor therapy in itself.
Collapse
Affiliation(s)
- Astrid E Visser
- Leiden University, Molecular Genetics, Leiden Institute of Chemistry, 2300 RA Leiden, The Netherlands
| | - Claude Backendorf
- Leiden University, Molecular Genetics, Leiden Institute of Chemistry, 2300 RA Leiden, The Netherlands
| | - Mathieu Noteborn
- Leiden University, Biological Chemistry, Leiden Institute of Chemistry, 2300 RA Leiden, The Netherlands
| |
Collapse
|
20
|
Ghosh R, Girigoswami K. NADH dehydrogenase subunits are overexpressed in cells exposed repeatedly to H2O2. Mutat Res 2007; 638:210-5. [PMID: 17905312 DOI: 10.1016/j.mrfmmm.2007.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Revised: 07/11/2007] [Accepted: 08/16/2007] [Indexed: 11/28/2022]
Abstract
Cells conditioned by repeated treatments with low doses of H(2)O(2,) were compared with its parental V79 cells for expression of ND1 and ND4 subunits of NADH dehydrogenase, a mitochondrial gene. It was found that ND1 and ND4 subunits were overexpressed in these conditioned cells. These cells were also found to be resistant to killing upon gamma-irradiation through suppression of apoptotic cell death. On irradiation, the expression of both subunits decreased in both cell types, but overall there was more expression of both subunits in the conditioned cells. These findings indicate alteration in the expression of NADH dehydrogenase, a mitochondrial gene, could be involved in the recovery of gamma-irradiated cells through inhibition of apoptosis.
Collapse
Affiliation(s)
- Rita Ghosh
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741235, Nadia, India.
| | | |
Collapse
|
21
|
Majumder P, Choudhury A, Banerjee M, Lahiri A, Bhattacharyya NP. Interactions of HIPPI, a molecular partner of Huntingtin interacting protein HIP1, with the specific motif present at the putative promoter sequence of the caspase-1, caspase-8 and caspase-10 genes. FEBS J 2007; 274:3886-99. [PMID: 17623017 DOI: 10.1111/j.1742-4658.2007.05922.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
To investigate the mechanism of increased expression of caspase-1 caused by exogenous Hippi, observed earlier in HeLa and Neuro2A cells, in this work we identified a specific motif AAAGACATG (- 101 to - 93) at the caspase-1 gene upstream sequence where HIPPI could bind. Various mutations in this specific sequence compromised the interaction, showing the specificity of the interactions. In the luciferase reporter assay, when the reporter gene was driven by caspase-1 gene upstream sequences (- 151 to - 92) with the mutation G to T at position - 98, luciferase activity was decreased significantly in green fluorescent protein-Hippi-expressing HeLa cells in comparison to that obtained with the wild-type caspase-1 gene 60 bp upstream sequence, indicating the biological significance of such binding. It was observed that the C-terminal 'pseudo' death effector domain of HIPPI interacted with the 60 bp (- 151 to - 92) upstream sequence of the caspase-1 gene containing the motif. We further observed that expression of caspase-8 and caspase-10 was increased in green fluorescent protein-Hippi-expressing HeLa cells. In addition, HIPPI interacted in vitro with putative promoter sequences of these genes, containing a similar motif. In summary, we identified a novel function of HIPPI; it binds to specific upstream sequences of the caspase-1, caspase-8 and caspase-10 genes and alters the expression of the genes. This result showed the motif-specific interaction of HIPPI with DNA, and indicates that it could act as transcription regulator.
Collapse
Affiliation(s)
- P Majumder
- Structural Genomics Section, Saha Institute of Nuclear Physics, Bidhan Nagar, Kolkata, India
| | | | | | | | | |
Collapse
|
22
|
Trushina E, McMurray CT. Oxidative stress and mitochondrial dysfunction in neurodegenerative diseases. Neuroscience 2007; 145:1233-48. [PMID: 17303344 DOI: 10.1016/j.neuroscience.2006.10.056] [Citation(s) in RCA: 323] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 10/24/2006] [Accepted: 10/27/2006] [Indexed: 11/16/2022]
Abstract
In recent years, it has become increasingly clear that mitochondrial dysfunction and oxidative damage are major contributors to neuronal loss. Free radicals, typically generated from mitochondrial respiration, cause oxidative damage of nucleic acids, lipids, carbohydrates and proteins. Despite enormous amount of effort, however, the mechanism by which oxidative damage causes neuronal death is not well understood. Emerging data from a number of neurodegenerative diseases suggest that there may be common features of toxicity that are related to oxidative damage. In this review, while focusing on Huntington's disease (HD), we discuss similarities among HD, Friedreich ataxia and xeroderma pigmentosum, which provide insight into shared mechanisms of neuronal death.
Collapse
Affiliation(s)
- E Trushina
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | |
Collapse
|
23
|
Majumder P, Chattopadhyay B, Sukanya S, Ray T, Banerjee M, Mukhopadhyay D, Bhattacharyya NP. Interaction of HIPPI with putative promoter sequence of caspase-1 in vitro and in vivo. Biochem Biophys Res Commun 2007; 353:80-5. [PMID: 17173859 DOI: 10.1016/j.bbrc.2006.11.138] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2006] [Accepted: 11/21/2006] [Indexed: 11/27/2022]
Abstract
To investigate the mechanism of increased expression of caspase-1 in Hippi expressing HeLa and Neuro 2A cells, reported earlier, we report here that HIPPI directly interacted with upstream sequence of caspase-1 gene (-700 to +17, 717 bp). Deletion of this 717 bp sequence and further analysis by electrophoretic mobility shift assay and fluorescence quenching revealed that HIPPI interacted with 60 bp (-151 to -92) upstream sequence of caspase-1. We also observed by chromatin immunoprecipitation assay that HIPPI interacted with the 717 bp sequence in vivo. In luciferase assay, when expression of the reporter gene was driven by either 717 bp or 60 bp caspase-1 upstream sequences, luciferase activity was increased in GFP-Hippi expressing HeLa cells in comparison to that obtained with parental HeLa cells with the same constructs. Similar result was obtained in Neuro2A cells with 717 bp caspase-1 upstream sequence. In summary, we showed that HIPPI could interact with the putative promoter sequence of caspase-1 and increased the expression of the downstream gene suggesting that HIPPI could act as transcription regulator.
Collapse
Affiliation(s)
- P Majumder
- Structural Genomics Section and Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, 1/AF Bidhan Nagar, Kolkata 700 064, India
| | | | | | | | | | | | | |
Collapse
|
24
|
Banerjee M, Majumder P, Bhattacharyya NP, Dattagupta JK, Sen U. Cloning, expression, purification, crystallization and preliminary crystallographic analysis of pseudo death-effector domain of HIPPI, a molecular partner of Huntingtin-interacting protein HIP-1. Acta Crystallogr Sect F Struct Biol Cryst Commun 2006; 62:1247-50. [PMID: 17142908 PMCID: PMC2225382 DOI: 10.1107/s1744309106046628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Accepted: 11/06/2006] [Indexed: 02/08/2023]
Abstract
The formation of a heterodimer between Huntingtin-interacting protein-1 (HIP-1) and its novel partner HIPPI (HIP-1 protein interactor) through their pseudo death-effector domains (pDEDs) is a key step that recruits caspase-8 and initiates apoptosis. This could be one of the pathways by which apoptosis is increased in Huntington's disease (HD). A construct consisting of the pDED of HIPPI has been cloned and overexpressed as 6NH-tagged protein and purified by Ni-NTA affinity chromatography. Crystals of the pDED of HIPPI were grown in space group P4(1), with unit-cell parameters a = b = 77.42, c = 33.31 A and a calculated Matthews coefficient of 1.88 A3 Da(-1) (33% solvent content) with two molecules per asymmetric unit.
Collapse
Affiliation(s)
- Manisha Banerjee
- Structural Genomics Section, Saha Institute of Nuclear Physics, 1/AF Bidhan Nagar, Kolkata 700064, India
| | - Pritha Majumder
- Structural Genomics Section, Saha Institute of Nuclear Physics, 1/AF Bidhan Nagar, Kolkata 700064, India
| | - Nitai P. Bhattacharyya
- Structural Genomics Section, Saha Institute of Nuclear Physics, 1/AF Bidhan Nagar, Kolkata 700064, India
| | - Jiban K. Dattagupta
- Structural Genomics Section, Saha Institute of Nuclear Physics, 1/AF Bidhan Nagar, Kolkata 700064, India
| | - Udayaditya Sen
- Structural Genomics Section, Saha Institute of Nuclear Physics, 1/AF Bidhan Nagar, Kolkata 700064, India
- Correspondence e-mail:
| |
Collapse
|