1
|
Chierichetti M, Cristofani R, Crippa V, Ferrari V, Cozzi M, Casarotto E, Pramaggiore P, Cornaggia L, Patelli G, Mohamed A, Piccolella M, Galbiati M, Rusmini P, Tedesco B, Poletti A. Small heat shock protein B8: from cell functions to its involvement in diseases and potential therapeutic applications. Neural Regen Res 2025; 20:2872-2886. [PMID: 39435632 PMCID: PMC11826450 DOI: 10.4103/nrr.nrr-d-24-00517] [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: 05/06/2024] [Revised: 08/05/2024] [Accepted: 08/29/2024] [Indexed: 10/23/2024] Open
Abstract
Heat shock protein family B (small) member 8 (HSPB8) is a 22 kDa ubiquitously expressed protein belonging to the family of small heat shock proteins. HSPB8 is involved in various cellular mechanisms mainly related to proteotoxic stress response and in other processes such as inflammation, cell division, and migration. HSPB8 binds misfolded clients to prevent their aggregation by assisting protein refolding or degradation through chaperone-assisted selective autophagy. In line with this function, the pro-degradative activity of HSPB8 has been found protective in several neurodegenerative and neuromuscular diseases characterized by protein misfolding and aggregation. In cancer, HSPB8 has a dual role being capable of exerting either a pro- or an anti-tumoral activity depending on the pathways and factors expressed by the model of cancer under investigation. Moreover, HSPB8 exerts a protective function in different diseases by modulating the inflammatory response, which characterizes not only neurodegenerative diseases, but also other chronic or acute conditions affecting the nervous system, such as multiple sclerosis and intracerebellar hemorrhage. Of note, HSPB8 modulation may represent a therapeutic approach in other neurological conditions that develop as a secondary consequence of other diseases. This is the case of cognitive impairment related to diabetes mellitus, in which HSPB8 exerts a protective activity by assuring mitochondrial homeostasis. This review aims to summarize the diverse and multiple functions of HSPB8 in different pathological conditions, focusing on the beneficial effects of its modulation. Drug-based and alternative therapeutic approaches targeting HSPB8 and its regulated pathways will be discussed, emphasizing how new strategies for cell and tissue-specific delivery represent an avenue to advance in disease treatments.
Collapse
Affiliation(s)
- Marta Chierichetti
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Riccardo Cristofani
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Valeria Crippa
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Veronica Ferrari
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Marta Cozzi
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Elena Casarotto
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Paola Pramaggiore
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Laura Cornaggia
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Guglielmo Patelli
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Ali Mohamed
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Margherita Piccolella
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Mariarita Galbiati
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Paola Rusmini
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Barbara Tedesco
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Angelo Poletti
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
2
|
Feng Y, Ma J, Bo Z, Yue D, Wang Y. The crucial role of small heat shock proteins in prostate cancer: mechanisms and new therapeutic perspectives. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2025; 1868:195090. [PMID: 40222452 DOI: 10.1016/j.bbagrm.2025.195090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
As resistance to new anti-androgen drugs occurs more frequently, increasing numbers of researchers are exploring alternative key molecular targets for prostate cancer treatment. The small heat shock protein (sHSP) family is a subclass of heat shock proteins (HSPs). Due to the smaller molecular size of their monomers, they often function as large oligomeric complexes with diverse biological roles, thus garnering increasing attention from urologists. Different members of the sHSP family exhibit distinct biological roles in prostate cancer, offering a new perspective for precision therapy. In this review, we summarize the specific roles of sHSP family members in prostate cancer and analyze their similarities and differences. Additionally, we discuss and review the drugs targeting various sHSPs in prostate cancer, providing new insights into the exploration and further application of sHSP-targeted therapies.
Collapse
Affiliation(s)
- Yuankang Feng
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Jialu Ma
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zhihao Bo
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Dan Yue
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300211, China.
| | - Yong Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
3
|
Adamiak K, Gaida VA, Schäfer J, Bosse L, Diemer C, Reiter RJ, Slominski AT, Steinbrink K, Sionkowska A, Kleszczyński K. Melatonin/Sericin Wound Healing Patches: Implications for Melanoma Therapy. Int J Mol Sci 2024; 25:4858. [PMID: 38732075 PMCID: PMC11084828 DOI: 10.3390/ijms25094858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Melatonin and sericin exhibit antioxidant properties and may be useful in topical wound healing patches by maintaining redox balance, cell integrity, and regulating the inflammatory response. In human skin, melatonin suppresses damage caused by ultraviolet radiation (UVR) which involves numerous mechanisms associated with reactive oxygen species/reactive nitrogen species (ROS/RNS) generation and enhancing apoptosis. Sericin is a protein mainly composed of glycine, serine, aspartic acid, and threonine amino acids removed from the silkworm cocoon (particularly Bombyx mori and other species). It is of interest because of its biodegradability, anti-oxidative, and anti-bacterial properties. Sericin inhibits tyrosinase activity and promotes cell proliferation that can be supportive and useful in melanoma treatment. In recent years, wound healing patches containing sericin and melatonin individually have attracted significant attention by the scientific community. In this review, we summarize the state of innovation of such patches during 2021-2023. To date, melatonin/sericin-polymer patches for application in post-operational wound healing treatment has been only sparingly investigated and it is an imperative to consider these materials as a promising approach targeting for skin tissue engineering or regenerative dermatology.
Collapse
Affiliation(s)
- Katarzyna Adamiak
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University, Gagarin 7, 87-100 Toruń, Poland; (K.A.); (A.S.)
| | - Vivian A. Gaida
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| | - Jasmin Schäfer
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| | - Lina Bosse
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| | - Clara Diemer
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health, San Antonio, TX 78229, USA;
| | - Andrzej T. Slominski
- Department of Dermatology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Pathology and Laboratory Medicine Service, VA Medical Center, Birmingham, AL 35294, USA
| | - Kerstin Steinbrink
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| | - Alina Sionkowska
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University, Gagarin 7, 87-100 Toruń, Poland; (K.A.); (A.S.)
| | - Konrad Kleszczyński
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| |
Collapse
|
4
|
Yang Y, Ma S, Ye Z, Zheng Y, Zheng Z, Liu X, Zhou X. Oncogenic DNA methyltransferase 1 activates the PI3K/AKT/mTOR signalling by blocking the binding of HSPB8 and BAG3 in melanoma. Epigenetics 2023; 18:2239607. [PMID: 37523636 PMCID: PMC10392740 DOI: 10.1080/15592294.2023.2239607] [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: 07/22/2022] [Revised: 02/27/2023] [Accepted: 04/07/2023] [Indexed: 08/02/2023] Open
Abstract
Abnormal DNA methylation has been observed in multiple malignancies, including melanoma. In this study, we initially noticed the overexpression of DNA methyltransferase 1 (DNMT1) in melanoma samples in bioinformatics analysis and, subsequently, validated it in the purchased melanoma cell lines. After treatment with short-hairpin RNAs or Decitabine (a DNA methylation inhibitor), silencing of DNMT1 was demonstrated to suppress cell viability and invasive and migratory potentials as well as to augment apoptosis and autophagy in melanoma cells. To further explore the downstream mechanisms, we revealed that DNMT1 inhibited HSPB8 expression through augmenting HSPB8 methylation, thereby suppressing the binding between HSPB8 and BAG3. Then, we elucidated through a series of gain- and loss- of function assays that the interplay of HSPB8 and BAG3 blocked the PI3K/AKT/mTOR pathway, thereby repressing the malignant phenotypes of melanoma cells and contributing to melanoma cell apoptosis and autophagy. We further established a mouse model of melanoma and substantiated that DNMT1 enhanced the in vivo tumorigenesis of melanoma cells via activation of the PI3K/AKT/mTOR pathway through repressing the binding between HSPB8 and BAG3. Taken together, our data supported that DNMT1 repressed the binding between HSPB8 and BAG3 and activated the PI3K/AKT/mTOR pathway, thus playing a tumour-promoting role in melanoma.
Collapse
Affiliation(s)
- Yemei Yang
- Department of Dermatology and Venerology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, P. R. China
| | - Shengfang Ma
- Department of Dermatology, Baoshihua Hospital of Gansu Province, Lanzhou, P. R. China
| | - Zi Ye
- College of Information and Sciences, The Pennsylvania State University, University of Pennsylvania, Philadelphia, USA
| | - Yushi Zheng
- Department of Dermatology and Venerology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, P. R. China
| | - Zhenjiong Zheng
- Department of Dermatology and Venerology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, P. R. China
| | - Xiongshan Liu
- Department of Dermatology and Venerology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, P. R. China
| | - Xianyi Zhou
- Department of Dermatology and Venerology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
5
|
Cristofani R, Piccolella M, Montagnani Marelli M, Tedesco B, Poletti A, Moretti RM. HSPB8 counteracts tumor activity of BRAF- and NRAS-mutant melanoma cells by modulation of RAS-prenylation and autophagy. Cell Death Dis 2022; 13:973. [PMID: 36400750 PMCID: PMC9674643 DOI: 10.1038/s41419-022-05365-9] [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: 05/10/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/19/2022]
Abstract
Cutaneous melanoma is one of the most aggressive and lethal forms of skin cancer. Some specific driver mutations have been described in multiple oncogenes including BRAF and NRAS that are mutated in 60-70% and 15-20% of melanoma, respectively. The aim of this study was to evaluate the role of Small Heat Shock Protein B8 (HSPB8) on cell growth and migration of both BLM (BRAFwt/NRASQ61R) and A375 (BRAFV600E/NRASwt) human melanoma cell lines. HSPB8 is a member of the HSPB family of chaperones involved in protein quality control (PQC) system and contributes to chaperone assisted selective autophagy (CASA) as well as in the regulation of mitotic spindle. In cancer, HSPB8 has anti- or pro-tumoral action depending on tumor type. In melanoma cell lines characterized by low HSPB8 levels, we demonstrated that the restoration of HSPB8 expression causes cell growth arrest, reversion of EMT (Epithelial-Mesenchymal Transition)-like phenotype switching and antimigratory effect, independently from the cell mutational status. We demonstrated that HSPB8 regulates the levels of the active prenylated form of NRAS in NRAS-mutant and NRAS-wild-type melanoma cell lines. Consequently, the inhibition of NRAS impairs the activation of Akt/mTOR pathway inducing autophagy activation. Autophagy can play a dual role in regulating cell death and survival. We have therefore demonstrated that HSPB8-induced autophagy is a crucial event that counteracts cell growth in melanoma. Collectively, our results suggest that HSPB8 has an antitumoral action in melanoma cells characterized by BRAF and NRAS mutations.
Collapse
Affiliation(s)
- Riccardo Cristofani
- grid.4708.b0000 0004 1757 2822Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Margherita Piccolella
- grid.4708.b0000 0004 1757 2822Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Marina Montagnani Marelli
- grid.4708.b0000 0004 1757 2822Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Barbara Tedesco
- grid.4708.b0000 0004 1757 2822Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy ,grid.417894.70000 0001 0707 5492Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Angelo Poletti
- grid.4708.b0000 0004 1757 2822Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Roberta Manuela Moretti
- grid.4708.b0000 0004 1757 2822Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
6
|
Cristofani R, Piccolella M, Crippa V, Tedesco B, Montagnani Marelli M, Poletti A, Moretti RM. The Role of HSPB8, a Component of the Chaperone-Assisted Selective Autophagy Machinery, in Cancer. Cells 2021; 10:335. [PMID: 33562660 PMCID: PMC7915307 DOI: 10.3390/cells10020335] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
The cellular response to cancer-induced stress is one of the major aspects regulating cancer development and progression. The Heat Shock Protein B8 (HSPB8) is a small chaperone involved in chaperone-assisted selective autophagy (CASA). CASA promotes the selective degradation of proteins to counteract cell stress such as tumor-induced stress. HSPB8 is also involved in (i) the cell division machinery regulating chromosome segregation and cell cycle arrest in the G0/G1 phase and (ii) inflammation regulating dendritic cell maturation and cytokine production. HSPB8 expression and role are tumor-specific, showing a dual and opposite role. Interestingly, HSPB8 may be involved in the acquisition of chemoresistance to drugs. Despite the fact the mechanisms of HSPB8-mediated CASA activation in tumors need further studies, HSPB8 could represent an important factor in cancer induction and progression and it may be a potential target for anticancer treatment in specific types of cancer. In this review, we will discuss the molecular mechanism underlying HSPB8 roles in normal and cancer conditions. The basic mechanisms involved in anti- and pro-tumoral activities of HSPB8 are deeply discussed together with the pathways that modulate HSPB8 expression, in order to outline molecules with a beneficial effect for cancer cell growth, migration, and death.
Collapse
|
7
|
Milani A, Basirnejad M, Bolhassani A. Heat-shock proteins in diagnosis and treatment: an overview of different biochemical and immunological functions. Immunotherapy 2020; 11:215-239. [PMID: 30730280 DOI: 10.2217/imt-2018-0105] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Heat-shock proteins (HSPs) have been involved in different functions including chaperone activity, protein folding, apoptosis, autophagy and immunity. The HSP families have powerful effects on the stimulation of innate immune responses through Toll-like receptors and scavenger receptors. Moreover, HSP-mediated phagocytosis directly enhances the processing and presentation of internalized antigens via the endocytic pathway in adaptive immune system. These properties of HSPs have been used for development of prophylactic and therapeutic vaccines against infectious and noninfectious diseases. Several studies also demonstrated the relationship between HSPs and drug resistance as well as their use as a novel biomarker for detecting tumors in patients. The present review describes different roles of HSPs in biology and medicine especially biochemical and immunological aspects.
Collapse
Affiliation(s)
- Alireza Milani
- Department of Hepatitis & AIDS, Pasteur Institute of Iran, Tehran, Iran.,Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | | | - Azam Bolhassani
- Department of Hepatitis & AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
8
|
Aurelian L, Balan I. GABA AR α2-activated neuroimmune signal controls binge drinking and impulsivity through regulation of the CCL2/CX3CL1 balance. Psychopharmacology (Berl) 2019; 236:3023-3043. [PMID: 31030249 DOI: 10.1007/s00213-019-05220-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/04/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND PURPOSE Toll-like receptors (TLRs) are a family of innate immune system receptors that respond to pathogen-derived and tissue damage-related ligands and are increasingly recognized for their impact on homeostasis and its dysregulation in the nervous system. TLR signaling participates in brain injury and addiction, but its role in the alcohol-seeking behavior, which initiates alcohol drinking, is still poorly understood. In this review, we discuss our findings designed to elucidate the potential contribution of the activated TLR4 signal located in neurons, on impulsivity and the predisposition to initiate alcohol drinking (binge drinking). RESULTS Our findings indicate that the TLR4 signal is innately activated in neurons from alcohol-preferring subjects, identifying a genetic contribution to the regulation of impulsivity and the alcohol-seeking propensity. Signal activation is through the non-canonical, previously unknown, binding of TLR4 to the α2 subunit of the γ-aminobutyric 2 acid A receptor (GABAAR α2). Activation is sustained by the stress hormone corticotrophin-releasing factor (CRF) and additional still poorly recognized ligand/scaffold proteins. Focus is on the effect of TLR4 signal activation on the balance between pro- and anti-inflammatory chemokines [chemokine (C-C motif) ligand 2 (CCL2)/chemokine (C-X3-C motif) ligand 1 (CX3CL1)] and its effect on binge drinking. CONCLUSION The results are discussed within the context of current findings on the distinct activation and functions of TLR signals located in neurons, as opposed to immune cells. They indicate that the balance between pro- and anti-inflammatory TLR4 signaling plays a major role in binge drinking. These findings have major impact on future basic and translational research, including the development of potential therapeutic and preventative strategies.
Collapse
Affiliation(s)
- Laure Aurelian
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Stanford University School of Medicine OFDD, Stanford, CA, 94305, USA.
| | - Irina Balan
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
9
|
Yao J, Chen Y, Nguyen DT, Thompson ZJ, Eroshkin AM, Nerlakanti N, Patel AK, Agarwal N, Teer JK, Dhillon J, Coppola D, Zhang J, Perera R, Kim Y, Mahajan K. The Homeobox gene, HOXB13, Regulates a Mitotic Protein-Kinase Interaction Network in Metastatic Prostate Cancers. Sci Rep 2019; 9:9715. [PMID: 31273254 PMCID: PMC6609629 DOI: 10.1038/s41598-019-46064-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 06/18/2019] [Indexed: 12/15/2022] Open
Abstract
HOXB13, a homeodomain transcription factor, is linked to recurrence following radical prostatectomy. While HOXB13 regulates Androgen Receptor (AR) functions in a context dependent manner, its critical effectors in prostate cancer (PC) metastasis remain largely unknown. To identify HOXB13 transcriptional targets in metastatic PCs, we performed integrative bioinformatics analysis of differentially expressed genes (DEGs) in the proximity of the human prostate tumor-specific AR binding sites. Unsupervised Principal Component Analysis (PCA) led to a focused core HOXB13 target gene-set referred to as HOTPAM9 (HOXB13 Targets separating Primary And Metastatic PCs). HOTPAM9 comprised 7 mitotic kinase genes overexpressed in metastatic PCs, TRPM8, and the heat shock protein HSPB8, whose levels were significantly lower in metastatic PCs compared to the primary disease. The expression of a two-gene set, CIT and HSPB8 with an overall balanced accuracy of 98.8% and a threshold value of 0.2347, was sufficient to classify metastasis. HSPB8 mRNA expression was significantly increased following HOXB13 depletion in multiple metastatic CRPC models. Increased expression of HSPB8 by the microtubule inhibitor Colchicine or by exogenous means suppressed migration of mCRPC cells. Collectively, our results indicate that HOXB13 promotes metastasis of PCs by coordinated regulation of mitotic kinases and blockade of a putative tumor suppressor gene.
Collapse
Affiliation(s)
- Jiqiang Yao
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Yunyun Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Duy T Nguyen
- Department of Surgery, Washington University in St. Louis, MO, USA
| | - Zachary J Thompson
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Alexey M Eroshkin
- Bioinformatics Core, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Niveditha Nerlakanti
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ami K Patel
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Neha Agarwal
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Jasreman Dhillon
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Domenico Coppola
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Jingsong Zhang
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ranjan Perera
- Analytical Genomics and Bioinformatics, Sanford Burnham Prebys Discovery Institute, Orlando, FL, USA
| | - Youngchul Kim
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Kiran Mahajan
- Department of Surgery, Washington University in St. Louis, MO, USA.
| |
Collapse
|
10
|
Exploring the multifaceted roles of heat shock protein B8 (HSPB8) in diseases. Eur J Cell Biol 2018; 97:216-229. [PMID: 29555102 DOI: 10.1016/j.ejcb.2018.03.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 02/06/2023] Open
Abstract
HSPB8 is a member of ubiquitous small heat shock protein (sHSP) family, whose expression is induced in response to a wide variety of unfavorable physiological and environmental conditions. Investigation of HSPB8 structure indicated that HSPB8 belongs to the group of so-called intrinsically disordered proteins and possesses a highly flexible structure. Unlike most other sHSPs, HSPB8 tends to form small-molecular-mass oligomers and exhibits substrate-dependent chaperone activity. In cooperation with BAG3, the chaperone activity of HSPB8 was reported to be involved in the delivery of misfolded proteins to the autophagy machinery. Through this way, HSPB8 interferes with pathological processes leading to neurodegenerative diseases. Accordingly, published studies have identified genetic links between mutations of HSPB8 and some kind of neuromuscular diseases, further supporting its important role in neurodegenerative disorders. In addition to their anti-aggregation properties, HSPB8 is indicated to interact with a wide range of client proteins, modulating their maturations and activities, and therefore, regulates a large repertoire of cellular functions, including apoptosis, proliferation, inflammation and etc. As a result, HSPB8 has key roles in cancer biology, autoimmune diseases, cardiac diseases and cerebral vascular diseases.
Collapse
|
11
|
De Munck J, Binks A, McNeish IA, Aerts JL. Oncolytic virus-induced cell death and immunity: a match made in heaven? J Leukoc Biol 2017; 102:631-643. [PMID: 28720686 DOI: 10.1189/jlb.5ru0117-040r] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/22/2017] [Accepted: 05/22/2017] [Indexed: 12/18/2022] Open
Abstract
Our understanding of the mechanisms responsible for cancer development has increased enormously over the last decades. However, for many cancers, this has not been translated into a significant improvement in overall survival, and overall mortality remains high. Treatment for many malignancies remains based on surgery, chemotherapy, and radiotherapy. Significant progress has been made toward the development of more specific, more potent, and less invasive treatment modalities, but such targeted therapies remain the exception for most cancers. Thus, cancer therapies based on a different mechanism of action should be explored. The immune system plays an important role in keeping tumor growth at bay. However, in many cases, these responses are not strong enough to keep tumor growth under control. Thus, immunotherapy aims to boost the immune system to suppress tumor growth efficiently. This has been demonstrated by the recent successes of immune checkpoint therapy in several cancers. Oncolytic viruses (OVs) are another exciting class of immunotherapy agent. As well as replicating selectively within and killing tumor cells, OVs are able to elicit potent anti-tumor immune responses. Therapeutic vaccination with OVs, also referred to as cancer virotherapy, can thus be tailored to elicit vigorous cellular immune responses and even target individual malignancies in a personalized manner. In this review, we will describe the intricate link among oncolytic virotherapy, tumor immunology, and immunogenic cell death (ICD) and discuss ways to harness optimally their potential for future cancer therapy.
Collapse
Affiliation(s)
- Jolien De Munck
- Laboratory for Pharmaceutical Biotechnology and Molecular Biology, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Alex Binks
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Iain A McNeish
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Joeri L Aerts
- Laboratory for Pharmaceutical Biotechnology and Molecular Biology, Vrije Universiteit Brussel, Brussels, Belgium; and
| |
Collapse
|
12
|
Micevic G, Theodosakis N, Bosenberg M. Aberrant DNA methylation in melanoma: biomarker and therapeutic opportunities. Clin Epigenetics 2017; 9:34. [PMID: 28396701 PMCID: PMC5381063 DOI: 10.1186/s13148-017-0332-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/24/2017] [Indexed: 12/18/2022] Open
Abstract
Aberrant DNA methylation is an epigenetic hallmark of melanoma, known to play important roles in melanoma formation and progression. Recent advances in genome-wide methylation methods have provided the means to identify differentially methylated genes, methylation signatures, and potential biomarkers. However, despite considerable effort and advances in cataloging methylation changes in melanoma, many questions remain unanswered. The aim of this review is to summarize recent developments, emerging trends, and important unresolved questions in the field of aberrant DNA methylation in melanoma. In addition to reviewing recent developments, we carefully synthesize the findings in an effort to provide a framework for understanding the current state and direction of the field. To facilitate clarity, we divided the review into DNA methylation changes in melanoma, biomarker opportunities, and therapeutic developments. We hope this review contributes to accelerating the utilization of the diagnostic, prognostic, and therapeutic potential of DNA methylation for the benefit of melanoma patients.
Collapse
Affiliation(s)
- Goran Micevic
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520 USA.,Department of Pathology, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Nicholas Theodosakis
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520 USA.,Department of Pathology, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Marcus Bosenberg
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520 USA.,Department of Pathology, Yale University School of Medicine, New Haven, CT 06520 USA
| |
Collapse
|
13
|
Aurelian L, Bollino D, Colunga A. The oncolytic virus ΔPK has multimodal anti-tumor activity. Pathog Dis 2016; 74:ftw050. [PMID: 27242376 DOI: 10.1093/femspd/ftw050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2016] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses (OVs) are an emerging cancer therapeutic, with a near complete absence of serious adverse effects. However, clinical efficacy is relatively modest, related to poor tumor penetration, failure to lyse cancer stem cells (CSCs) and blockade of immunogenic cell death by the immunosuppressive tumor microenvironment. To overcome such limitations, we developed an OV (known as ΔPK) with multimodal anti-tumor activity. ΔPK has potent anti-tumor activity both in melanoma cell lines and xenograft animal models, associated with virus replication and the induction of multiple independent programmed cell death pathways. It lyses CSCs through autophagy modulation and it reverses the immunosuppressive tumor microenvironment by altering the balance of cytokines secreted by the tumor cells. This includes decreased tumor cell secretion of the immunosuppressive and procancerous cytokines IL-10 and IL-18 and concomitant increased secretion of the proinflammatory cytokines TNF-α, GM-CSF, IL-6 and IL-1β. ΔPK also upregulates the NKG2D ligand, MICA expressed by cytotoxic NK and T cells, and downregulates the negative immune checkpoint regulator cytotoxic T-lymphocyte antigen-4 (CTLA-4). ΔPK is well tolerated in human patients in whom it also alters the Th1/Th2 balance. Further studies are designed to elucidate the role of these contributions in different tumor types.
Collapse
Affiliation(s)
- Laure Aurelian
- Department of Microbiology and Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dominique Bollino
- Department of Microbiology and Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Aric Colunga
- Department of Microbiology and Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
14
|
Jiang Z, Harrington P, Zhang M, Marjani SL, Park J, Kuo L, Pribenszky C, Tian X(C. Effects of High Hydrostatic Pressure on Expression Profiles of In Vitro Produced Vitrified Bovine Blastocysts. Sci Rep 2016; 6:21215. [PMID: 26883277 PMCID: PMC4756375 DOI: 10.1038/srep21215] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/19/2016] [Indexed: 11/10/2022] Open
Abstract
High hydrostatic pressure (HHP) has been used to pre-condition embryos before essential, yet potentially detrimental procedures such as cryopreservation. However, the mechanisms for HHP are poorly understood. We treated bovine blastocysts with three different HHP (40, 60 and 80 MPa) in combination with three recovery periods (0, 1 h, 2 h post HHP). Re-expansion rates were significantly higher at 40 and 60 but lower at 80 MPa after vitrification-warming in the treated groups than controls. Microarray analysis revealed 399 differentially expressed transcripts, representing 254 unique genes, among different groups. Gene ontology analysis indicated that HHP at 40 and 60 MPa promoted embryo competence through down-regulation of genes in cell death and apoptosis, and up-regulation of genes in RNA processing, cellular growth and proliferation. In contrast, 80 MPa up-regulated genes in apoptosis, and down-regulated protein folding and cell cycle-related genes. Moreover, gene expression was also influenced by the length of the recovery time after HHP. The significantly over-represented categories were apoptosis and cell death in the 1 h group, and protein folding, response to unfolded protein and cell cycle in the 2 h group compared to 0 h. Taken together, HHP promotes competence of vitrified bovine blastocysts through modest transcriptional changes.
Collapse
Affiliation(s)
- Zongliang Jiang
- Center for Regenerative Biology, Department of Animal Science, University of Connecticcut, Storrs, Connecticut, 06269, USA
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China, 510005
| | - Patrick Harrington
- Department of Statistics, University of Connecticut, Storrs, Connecticut, 06269, USA
| | - Ming Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China, 510005
| | - Sadie L. Marjani
- Center for Regenerative Biology, Department of Animal Science, University of Connecticcut, Storrs, Connecticut, 06269, USA
- Department of Biology, Central Connecticut State University, New Britain, Connecticut 06050, USA
| | - Joonghoon Park
- Center for Regenerative Biology, Department of Animal Science, University of Connecticcut, Storrs, Connecticut, 06269, USA
| | - Lynn Kuo
- Department of Statistics, University of Connecticut, Storrs, Connecticut, 06269, USA
| | - Csaba Pribenszky
- Laboratory of Assisted Reproduction, Department of Herd Health, University of Veterinary Science, Istvan u. 2. 1078 Budapest, Hungary
| | - Xiuchun (Cindy) Tian
- Center for Regenerative Biology, Department of Animal Science, University of Connecticcut, Storrs, Connecticut, 06269, USA
| |
Collapse
|
15
|
Arrigo AP, Ducarouge B, Lavial F, Gibert B. Immense Cellular Implications Associated to Small Stress Proteins Expression: Impacts on Human Pathologies. HEAT SHOCK PROTEINS 2015. [DOI: 10.1007/978-3-319-16077-1_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
16
|
Luo C, Qu H, Wang J, Wang Y, Ma J, Li C, Yang C, Hu X, Li N, Shu D. Genetic parameters and genome-wide association study of hyperpigmentation of the visceral peritoneum in chickens. BMC Genomics 2013; 14:334. [PMID: 23679099 PMCID: PMC3663821 DOI: 10.1186/1471-2164-14-334] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 05/07/2013] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Hyperpigmentation of the visceral peritoneum (HVP) has recently garnered much attention in the poultry industry because of the possible risk to the health of affected animals and the damage it causes to the appearance of commercial chicken carcasses. However, the heritable characters of HVP remain unclear. The objective of this study was to investigate the genetic parameters of HVP by genome-wide association study (GWAS) in chickens. RESULTS HVP was found to be influenced by genetic factors, with a heritability score of 0.33. HVP had positive genetic correlations with growth and carcass traits, such as leg muscle weight (rg = 0.34), but had negative genetic correlations with immune traits, such as the antibody response to Newcastle disease virus (rg = -0.42). The GWAS for HVP using 39,833 single nucleotide polymorphisms indicated the genetic factors associated with HVP displayed an additive effect rather than a dominance effect. In addition, we determined that three genomic regions, involving the 50.5-54.0 Mb region of chicken (Gallus gallus) chromosome 1 (GGA1), the 58.5-60.5 Mb region of GGA1, and the 10.5-12.0 Mb region of GGA20, were strongly associated (P < 6.28 × 10-7) with HVP in chickens. Variants in these regions explained >50% of additive genetic variance for HVP. This study also confirmed that expression of BMP7, which codes for a bone morphogenetic protein and is located in one of the candidate regions, was significantly higher in the visceral peritoneum of Huiyang Beard chickens with HVP than in that of chickens without pigmentation (P < 0.05). CONCLUSIONS HVP is a quantitative trait with moderate heritability. Genomic variants resulting in HVP were identified on GGA1 and GGA20, and expression of the BMP7 gene appears to be upregulated in HVP-affected chickens. Findings from this study should be used as a basis for further functional validation of candidate genes involved in HVP.
Collapse
Affiliation(s)
- Chenglong Luo
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, 1 Dafeng 1st Street, Wushan, Tianhe District, Guangzhou, Guangdong, 510640, China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, 510640, China
| | - Hao Qu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, 1 Dafeng 1st Street, Wushan, Tianhe District, Guangzhou, Guangdong, 510640, China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, 510640, China
| | - Jie Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, 1 Dafeng 1st Street, Wushan, Tianhe District, Guangzhou, Guangdong, 510640, China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, 510640, China
| | - Yan Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, 1 Dafeng 1st Street, Wushan, Tianhe District, Guangzhou, Guangdong, 510640, China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, 510640, China
| | - Jie Ma
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, 1 Dafeng 1st Street, Wushan, Tianhe District, Guangzhou, Guangdong, 510640, China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, 510640, China
| | - Chunyu Li
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, 1 Dafeng 1st Street, Wushan, Tianhe District, Guangzhou, Guangdong, 510640, China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, 510640, China
| | - Chunfen Yang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, 1 Dafeng 1st Street, Wushan, Tianhe District, Guangzhou, Guangdong, 510640, China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, 510640, China
| | - Xiaoxiang Hu
- State Key Laboratory for Agro-Biotechnology, China Agricultural University, Beijing, 100193, China
| | - Ning Li
- State Key Laboratory for Agro-Biotechnology, China Agricultural University, Beijing, 100193, China
| | - Dingming Shu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, 1 Dafeng 1st Street, Wushan, Tianhe District, Guangzhou, Guangdong, 510640, China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, 510640, China
| |
Collapse
|
17
|
Wen J, Hu Y, Luo KJ, Yang H, Zhang SS, Fu JH. Positive transforming growth factor-β activated kinase-1 expression has an unfavorable impact on survival in T3N1-3M0 esophageal squamous cell carcinomas. Ann Thorac Surg 2012; 95:285-90. [PMID: 23272845 DOI: 10.1016/j.athoracsur.2012.09.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 09/13/2012] [Accepted: 09/17/2012] [Indexed: 01/15/2023]
Abstract
BACKGROUND Transforming growth factor-β activated kinase-1 (TAK1) is a serine/threonine kinase in the mitogen-activated protein kinase kinase family. Previous studies have reported the role of TAK1 in cancer occurrence and progression; however, its role and clinical significance in esophageal squamous cell carcinoma (ESCC) has not been elucidated. We investigated the correlation of TAK1 expression and clinical outcome in T3N1-3M0 surgically resected ESCCs. METHODS Tissue microarrays constructed of 234 surgically resected T3N1-3M0 ESCC primary tumors were used for TAK1 evaluation by immunohistochemistry. The clinical and prognostic significance of TAK1 expression was analyzed statistically. RESULTS Positive expression of TAK1 was observed in 38.5% (90 of 234). The expression level of TAK1 in ESCCs at stage N2-3 was significantly higher than the expression level in those at stage N1 (p = 0.041). Patients with negative TAK1 expression demonstrated better overall survival than those with positive expression (median, 22.7 vs 17.4 months; p = 0.023). Multivariate analysis showed that TAK1 expression was an independent prognostic factor in ESCC (relative risk, 1.429; p = 0.021). CONCLUSIONS TAK1 expression correlates with lymph node metastasis and is a negative, independent prognostic factor in resected T3N1-3M0 ESCCs.
Collapse
Affiliation(s)
- Jing Wen
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | | | | | | | | | | |
Collapse
|
18
|
H11/HspB8 and Its Herpes Simplex Virus Type 2 Homologue ICP10PK Share Functions That Regulate Cell Life/Death Decisions and Human Disease. Autoimmune Dis 2012; 2012:395329. [PMID: 23056924 PMCID: PMC3463903 DOI: 10.1155/2012/395329] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/20/2012] [Accepted: 08/21/2012] [Indexed: 12/24/2022] Open
Abstract
Small heat shock proteins (sHsp) also known as HspB are a large family of widely expressed proteins that contain a 90 residues domain known as α-crystallin. Here, we focus on the family member H11/HspB8 and its herpes simplex virus type 2 (HSV-2) homologue ICP10PK, and discuss the possible impact of this relationship on human disease. H11/HspB8 and ICP10PK are atypical protein kinases. They share multi-functional activity that encompasses signaling, unfolded protein response (UPR) and the regulation of life cycle potential. In melanocytes H11/HspB8 causes growth arrest. It is silenced in a high proportion of melanoma prostate cancer, Ewing's sarcoma and hematologic malignancies through aberrant DNA methylation. Its restored expression induces cell death and inhibits tumor growth in xenograft models, identifying H11/HspB8 as a tumor suppressor. This function involves the activation of multiple and distinct death pathways, all of which initiate with H11/HspB8-mediated phosphorylation of transforming growth factor β-activated kinase 1 (TAK1). Both ICP10PK and H11/HspB8 were implicated in inflammatory processes that involve dendritic cells activation through Toll-like receptor-dependent pathways and may contribute to the onset of autoimmunity. The potential evolutionary relationship of H11/HspB8 to ICP10PK, its impact on human disorders and the development of therapeutic strategies are discussed.
Collapse
|
19
|
Restored expression of the atypical heat shock protein H11/HspB8 inhibits the growth of genetically diverse melanoma tumors through activation of novel TAK1-dependent death pathways. Cell Death Dis 2012; 3:e371. [PMID: 22898869 PMCID: PMC3434666 DOI: 10.1038/cddis.2012.108] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Melanoma is an aggressive and drug-resistant cancer in need of improved therapeutic strategies. Restored expression of transcriptionally silenced genes is a potential approach, but it is limited by the genetic diversity of the melanoma tumors. The atypical heat shock protein H11/HspB8 has kinase activity and is silenced in melanoma through aberrant DNA methylation. We report that its restored expression induces the death of genetically diverse melanoma lines and inhibits tumor growth through the activation of novel TAK1-dependent death pathways. These include (i) caspase-1 activation independent of the inflammasome through upregulation of apoptosis-associated speck-like protein containing a CARD (ASC), (ii) Beclin-1 upregulation through phosphorylation of mammalian target of rapamycin (mTOR) at S2481 and (iii) apoptosis caused by caspase-1-mediated Beclin-1 cleavage. These data extend current understanding of cell death-associated functions, underscore the strong therapeutic promise of H11/HspB8 and identify TAK1 as a potential intervention target in melanoma.
Collapse
|
20
|
Bucci C, Bakke O, Progida C. Charcot-Marie-Tooth disease and intracellular traffic. Prog Neurobiol 2012; 99:191-225. [PMID: 22465036 PMCID: PMC3514635 DOI: 10.1016/j.pneurobio.2012.03.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Revised: 12/23/2011] [Accepted: 03/13/2012] [Indexed: 12/23/2022]
Abstract
Mutations of genes whose primary function is the regulation of membrane traffic are increasingly being identified as the underlying causes of various important human disorders. Intriguingly, mutations in ubiquitously expressed membrane traffic genes often lead to cell type- or organ-specific disorders. This is particularly true for neuronal diseases, identifying the nervous system as the most sensitive tissue to alterations of membrane traffic. Charcot-Marie-Tooth (CMT) disease is one of the most common inherited peripheral neuropathies. It is also known as hereditary motor and sensory neuropathy (HMSN), which comprises a group of disorders specifically affecting peripheral nerves. This peripheral neuropathy, highly heterogeneous both clinically and genetically, is characterized by a slowly progressive degeneration of the muscle of the foot, lower leg, hand and forearm, accompanied by sensory loss in the toes, fingers and limbs. More than 30 genes have been identified as targets of mutations that cause CMT neuropathy. A number of these genes encode proteins directly or indirectly involved in the regulation of intracellular traffic. Indeed, the list of genes linked to CMT disease includes genes important for vesicle formation, phosphoinositide metabolism, lysosomal degradation, mitochondrial fission and fusion, and also genes encoding endosomal and cytoskeletal proteins. This review focuses on the link between intracellular transport and CMT disease, highlighting the molecular mechanisms that underlie the different forms of this peripheral neuropathy and discussing the pathophysiological impact of membrane transport genetic defects as well as possible future ways to counteract these defects.
Collapse
Affiliation(s)
- Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni, 73100 Lecce, Italy.
| | | | | |
Collapse
|
21
|
Cui XY, Wang N, Yang BX, Gao WF, Lin YM, Yao XR, Ma XT. HSPB8 is methylated in hematopoietic malignancies and overexpression of HSPB8 exhibits antileukemia effect. Exp Hematol 2011; 40:14-21. [PMID: 21914495 DOI: 10.1016/j.exphem.2011.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 08/29/2011] [Accepted: 09/07/2011] [Indexed: 12/20/2022]
Abstract
HSPB8 has been shown to be involved in regulation of cell proliferation and apoptosis, and it has also been found to have divergent properties in solid tumors. The purpose of this study was to investigate the expression and function of HSPB8 in hematopoietic malignancies. Expression and induced expression of HSPB8 was evaluated in hematopoietic tumor cell lines and bone marrow samples from patients with leukemia. Methylation status was investigated by methylation-specific polymerase chain reaction. The role of HSPB8 in hematopoietic malignancies was addressed by reintroducing HSPB8 expression into the K562 (leukemia) and Namalwa (lymphoma) cell lines. Expression of HSPB8 was absent in hematopoietic tumor cell lines and primary patient and normal volunteer samples. Promoter DNA methylation of HSPB8 was observed in these cells. HSPB8 expression could be restored after demethylation treatment with 5-aza-2'-deoxycytidine. Overexpression of HSPB8 reduced colony formation of both K562 and Namalwa cell lines, inhibited the cell growth of Namalwa in vitro, and suppressed tumor formation of K562 cells in vivo. The present study demonstrates that HSPB8 is silenced by DNA methylation in hematopoietic malignant and normal cells and its expression can be induced by treatment with 5-aza-2'-deoxycytidine. Overexpression of HSPB8 may have an antitumor activity in chronic myelogenous leukemia and lymphoma.
Collapse
Affiliation(s)
- Xue-Ying Cui
- State Key Laboratory for Experimental Hematology, Institute of Hematology, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin, China
| | | | | | | | | | | | | |
Collapse
|
22
|
Smith CC, Li B, Liu J, Lee KS, Aurelian L. The Levels of H11/HspB8 DNA methylation in human melanoma tissues and xenografts are a critical molecular marker for 5-Aza-2'-deoxycytidine therapy. Cancer Invest 2011; 29:383-95. [PMID: 21649464 PMCID: PMC3111925 DOI: 10.3109/07357907.2011.584588] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
H11/HspB8 is a functionally distinct small heat shock protein. It causes growth arrest in melanocytes, associated with the inhibition of Cyclin E/Cdk2 and β-catenin phosphorylation at the transcriptional activity site Ser(552) and is silenced through DNA methylation in 27/35 (77%) melanoma tissues/early cultures. 5-Aza-2'-deoxycytidine (Aza-C) induces melanoma cell death correlated with the levels of H11/HspB8 DNA methylation (p < .001). In line with low/moderate H11/HspB8 methylation, PI3-K inhibition increases Aza-C-induced cell death. Aza-C inhibits the growth of melanoma xenografts related to the levels of H11/HspB8 methylation, and a nonmethylated/non-TAK1 binding H11/HspB8 mutant confers Aza-C resistance. H11/HspB8 is a potential molecular marker for demethylation therapies.
Collapse
Affiliation(s)
- Cynthia C. Smith
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, 655 West Baltimore Street, Baltimore, Maryland 21201, USA
| | - Baiquan Li
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, 655 West Baltimore Street, Baltimore, Maryland 21201, USA
| | - Juan Liu
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, 655 West Baltimore Street, Baltimore, Maryland 21201, USA
| | - Kie-Sok Lee
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, 655 West Baltimore Street, Baltimore, Maryland 21201, USA
| | - Laure Aurelian
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, 655 West Baltimore Street, Baltimore, Maryland 21201, USA
| |
Collapse
|
23
|
High-throughput ectopic expression screen for tamoxifen resistance identifies an atypical kinase that blocks autophagy. Proc Natl Acad Sci U S A 2011; 108:2058-63. [PMID: 21233418 DOI: 10.1073/pnas.1018157108] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Resistance to tamoxifen in breast cancer patients is a serious therapeutic problem and major efforts are underway to understand underlying mechanisms. Resistance can be either intrinsic or acquired. We derived a series of subcloned MCF7 cell lines that were either highly sensitive or naturally resistant to tamoxifen and studied the factors that lead to drug resistance. Gene-expression studies revealed a signature of 67 genes that differentially respond to tamoxifen in sensitive vs. resistant subclones, which also predicts disease-free survival in tamoxifen-treated patients. High-throughput cell-based screens, in which >500 human kinases were independently ectopically expressed, identified 31 kinases that conferred drug resistance on sensitive cells. One of these, HSPB8, was also in the expression signature and, by itself, predicted poor clinical outcome in one cohort of patients. Further studies revealed that HSPB8 protected MCF7 cells from tamoxifen and blocked autophagy. Moreover, silencing HSBP8 induced autophagy and caused cell death. Tamoxifen itself induced autophagy in sensitive cells but not in resistant ones, and tamoxifen-resistant cells were sensitive to the induction of autophagy by other drugs. These results may point to an important role for autophagy in the sensitivity to tamoxifen.
Collapse
|
24
|
Cassar L, Li H, Jiang FX, Liu JP. TGF-beta induces telomerase-dependent pancreatic tumor cell cycle arrest. Mol Cell Endocrinol 2010; 320:97-105. [PMID: 20138964 DOI: 10.1016/j.mce.2010.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 01/27/2010] [Accepted: 02/02/2010] [Indexed: 12/21/2022]
Abstract
Recent studies suggest that transforming growth factor beta (TGF-beta) inhibits telomerase activity by repression of the telomerase reverse transcriptase (TERT) gene. In this report, we show that TGF-beta induces TERT repression-dependent apoptosis in pancreatic tumor, vascular smooth muscle, and cervical cancer cell cultures. TGF-beta activates Smad3 signaling, induces TERT gene repression and results in G1/S phase cell cycle arrest and apoptosis. TERT over-expression stimulates the G1/S phase transition and alienates TGF-beta-induced cell cycle arrest and apoptosis. Our data suggest that telomere maintenance is a limiting factor of the transition of the cell cycle. TGF-beta triggers cell cycle arrest and death by a mechanism involving telomerase deregulation of telomere maintenance.
Collapse
Affiliation(s)
- Lucy Cassar
- Department of Immunology, Monash University, Central Clinical School, AMREP, Commercial Road, Melbourne, Victoria 3004, Australia.
| | | | | | | |
Collapse
|
25
|
The HSV-2 mutant DeltaPK induces melanoma oncolysis through nonredundant death programs and associated with autophagy and pyroptosis proteins. Gene Ther 2009; 17:315-27. [PMID: 19798049 DOI: 10.1038/gt.2009.126] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Malignant melanoma is a highly aggressive and drug-resistant cancer. Virotherapy is a novel therapeutic strategy based on cancer cell lysis through selective virus replication. However, its clinical efficacy is modest, apparently related to poor virus replication within the tumors. We report that the growth compromised herpes simplex virus type 2 (HSV-2) mutant, DeltaPK, has strong oncolytic activity for melanoma largely caused by a mechanism other than replication-induced cell lysis. The ratio of dead cells (determined by trypan blue or ethidium homodimer staining) to cells that stain with antibody to the major capsid protein VP5 (indicative of productive infection) was 1.8-4.1 for different melanoma cultures at 24-72 h post-infection. Cell death was due to activation of calpain as well as caspases-7 and -3 and it was abolished by the combination of calpain (PD150606) and pancaspase (benzyloxycarbonyl-Val-Ala-Asp-fluormethyl ketone, z-VAD-fmk) inhibitors. Upregulation of the autopahgy protein Beclin-1 and the pro-apoptotic protein H11/HspB8 accompanied DeltaPK-induced melanoma oncolysis. Intratumoral DeltaPK injection (10(6)-10(7) plaque-forming unit (pfu)) significantly reduced melanoma tumor burden associated with calpain and caspases-7 and -3 activation, Beclin-1 and H11/HspB8 upregulation and activation of caspase-1-related inflammation. Complete remission was seen for 87.5% of the LM melanoma xenografts at 5 months after treatment termination. The data indicate that DeltaPK is a promising virotherapy for melanoma that functions through virus-induced programmed cell death pathways.
Collapse
|
26
|
Sui X, Li D, Qiu H, Gaussin V, Depre C. Activation of the Bone Morphogenetic Protein Receptor by H11Kinase/Hsp22 Promotes Cardiac Cell Growth and Survival. Circ Res 2009; 104:887-95. [DOI: 10.1161/circresaha.108.192328] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Xiangzhen Sui
- From the Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry New Jersey, New Jersey Medical School, Newark
| | - Dan Li
- From the Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry New Jersey, New Jersey Medical School, Newark
| | - Hongyu Qiu
- From the Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry New Jersey, New Jersey Medical School, Newark
| | - Vinciane Gaussin
- From the Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry New Jersey, New Jersey Medical School, Newark
| | - Christophe Depre
- From the Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry New Jersey, New Jersey Medical School, Newark
| |
Collapse
|