1
|
Yi K, Sun C, Yuan Y, Luo Z, Luo H, Xie Y. A new weapon: the application of tumor vaccines based on extracellular exosomal heat shock proteins in immunotherapy. Front Immunol 2025; 16:1510650. [PMID: 39911383 PMCID: PMC11794256 DOI: 10.3389/fimmu.2025.1510650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025] Open
Abstract
Despite the significant advancements in cancer research, innovative approaches are still needed to reduce tumor incidence, progression, and dissemination, as well as for prolonging patient survival. Currently, the development of cancer vaccines is gaining attention as a novel preventative and therapeutic strategy. Although the concept of cancer vaccination is not new, a limited number of vaccines have received approval for tumor therapy. Heat shock protein (HSP)-based vaccination represents a promising strategy that harnesses specific tumor antigens to activate immune responses. Exosomes (Exs) are highly heterogeneous bilayer vesicles capable of transporting various types of molecules through extracellular space. Compared with conventional anticancer drugs, exosomes exhibit low toxicity and good biocompatibility, and they can stimulate the immune system either directly or indirectly. Ex-based vaccines may elicit an antitumor immune response that generates memory cells capable of recognizing cancer antigens, thereby inhibiting disease progression. This paper reviews the potential applications of HSPs and exosomes in the prevention and treatment of solid tumors. Finally, we discuss the advantages of the extracellular exosomal heat shock protein (HSP-Ex) vaccine and future research directions aimed at optimizing heat shock protein-based cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Kexin Yi
- The Second Clinical Medical College, Nanchang University, Nanchang, China
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Chengpeng Sun
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yalin Yuan
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhaowei Luo
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongliang Luo
- Department of General Surgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
| | - Yunhe Xie
- The Second Clinical Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
2
|
Li Y, Cao S, Li Y. Mechanistic study of heat shock protein 60-mediated apoptosis in DF-1 cells. Poult Sci 2024; 103:103619. [PMID: 38603929 PMCID: PMC11017055 DOI: 10.1016/j.psj.2024.103619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/18/2024] [Accepted: 03/01/2024] [Indexed: 04/13/2024] Open
Abstract
Heat shock proteins (HSP) are a group of highly conserved molecular chaperones found in various organisms and have been associated with tumorigenesis, tumor progression, and metastasis. However, the relationship between HSP60 and apoptosis remains elusive. The aim of this study was to explore the role and regulatory mechanisms of apoptosis in response to altered HSP60 expression. We generated DF-1 cell lines of both HSP60 overexpression and knockdown and assessed their impact on apoptosis levels using ELISA and flow cytometry analyses. Additionally, we examined the transcription and protein expression levels of apoptosis-related signaling factors using fluorescence quantitative PCR (qPCR) and Western blotting analyses. Heat shock proteins 60 overexpression led to a significant decrease in apoptosis levels in DF-1 cells, which could be attributed to the downregulation of BAX and BAK expression, the upregulation of Bcl-2, and the decreased expression of Caspase 3. Conversely, HSP60 knockdown led to a substantial increase in apoptosis levels in DF-1 cells, facilitated by the downregulation of BAX and Bcl-2 expression, and the upregulation of BAK expression, which increased Caspase 3 levels, thereby promoting apoptosis. The findings of our study provide the first evidence of the inhibitory effect of HSP60 on apoptosis in DF-1 cells. These observations have significant implications for disease progression and cancer research, with potential medical applications.
Collapse
Affiliation(s)
- Yanlan Li
- College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, Shandong 252059, China
| | - Shengliang Cao
- College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, Shandong 252059, China
| | - Yubao Li
- College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, Shandong 252059, China.
| |
Collapse
|
3
|
Meng L, Zhang C, Yu P. Treating cancer through modulating exosomal protein loading and function: The prospects of natural products and traditional Chinese medicine. Pharmacol Res 2024; 203:107179. [PMID: 38615876 DOI: 10.1016/j.phrs.2024.107179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/22/2024] [Accepted: 04/05/2024] [Indexed: 04/16/2024]
Abstract
Exosomes, small yet vital extracellular vesicles, play an integral role in intercellular communication. They transport critical components, such as proteins, lipid bilayers, DNA, RNA, and glycans, to target cells. These vesicles are crucial in modulating the extracellular matrix and orchestrating signal transduction processes. In oncology, exosomes are pivotal in tumor growth, metastasis, drug resistance, and immune modulation within the tumor microenvironment. Exosomal proteins, noted for their stability and specificity, have garnered widespread attention. This review delves into the mechanisms of exosomal protein loading and their impact on tumor development, with a focus on the regulatory effects of natural products and traditional Chinese medicine on exosomal protein loading and function. These insights not only offer new strategies and methodologies for cancer treatment but also provide scientific bases and directions for future clinical applications.
Collapse
Affiliation(s)
- Lulu Meng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Pei Yu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
4
|
Kunachowicz D, Król-Kulikowska M, Raczycka W, Sleziak J, Błażejewska M, Kulbacka J. Heat Shock Proteins, a Double-Edged Sword: Significance in Cancer Progression, Chemotherapy Resistance and Novel Therapeutic Perspectives. Cancers (Basel) 2024; 16:1500. [PMID: 38672583 PMCID: PMC11048091 DOI: 10.3390/cancers16081500] [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: 03/19/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Heat shock proteins (Hsps) are involved in one of the adaptive mechanisms protecting cells against environmental and metabolic stress. Moreover, the large role of these proteins in the carcinogenesis process, as well as in chemoresistance, was noticed. This review aims to draw attention to the possibilities of using Hsps in developing new cancer therapy methods, as well as to indicate directions for future research on this topic. In order to discuss this matter, a thorough review of the latest scientific literature was carried out, taking into account the importance of selected proteins from the Hsp family, including Hsp27, Hsp40, Hsp60, Hsp70, Hsp90 and Hsp110. One of the more characteristic features of all Hsps is that they play a multifaceted role in cancer progression, which makes them an obvious target for modern anticancer therapy. Some researchers emphasize the importance of directly inhibiting the action of these proteins. In turn, others point to their possible use in the design of cancer vaccines, which would work by inducing an immune response in various types of cancer. Due to these possibilities, it is believed that the use of Hsps may contribute to the progress of oncoimmunology, and thus help in the development of modern anticancer therapies, which would be characterized by higher effectiveness and lower toxicity to the patients.
Collapse
Affiliation(s)
- Dominika Kunachowicz
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (D.K.); (M.K.-K.)
| | - Magdalena Król-Kulikowska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (D.K.); (M.K.-K.)
| | - Wiktoria Raczycka
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.R.); (J.S.); (M.B.)
| | - Jakub Sleziak
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.R.); (J.S.); (M.B.)
| | - Marta Błażejewska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.R.); (J.S.); (M.B.)
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine Santariškių g. 5, LT-08406 Vilnius, Lithuania
- DIVE IN AI, 53-307 Wroclaw, Poland
| |
Collapse
|
5
|
Virtuoso A, D’Amico G, Scalia F, De Luca C, Papa M, Maugeri G, D’Agata V, Caruso Bavisotto C, D’Amico AG. The Interplay between Glioblastoma Cells and Tumor Microenvironment: New Perspectives for Early Diagnosis and Targeted Cancer Therapy. Brain Sci 2024; 14:331. [PMID: 38671983 PMCID: PMC11048111 DOI: 10.3390/brainsci14040331] [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: 03/17/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) stands out as the most tremendous brain tumor, constituting 60% of primary brain cancers, accompanied by dismal survival rates. Despite advancements in research, therapeutic options remain limited to chemotherapy and surgery. GBM molecular heterogeneity, the intricate interaction with the tumor microenvironment (TME), and non-selective treatments contribute to the neoplastic relapse. Diagnostic challenges arise from GBM advanced-stage detection, necessitating the exploration of novel biomarkers for early diagnosis. Using data from the literature and a bioinformatic tool, the current manuscript delineates the molecular interplay between human GBM, astrocytes, and myeloid cells, underscoring selected protein pathways belonging to astroglia and myeloid lineage, which can be considered for targeted therapies. Moreover, the pivotal role of extracellular vesicles (EVs) in orchestrating a favorable microenvironment for cancer progression is highlighted, suggesting their utility in identifying biomarkers for GBM early diagnosis.
Collapse
Affiliation(s)
- Assunta Virtuoso
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Giuseppa D’Amico
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
| | - Federica Scalia
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
| | - Ciro De Luca
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (V.D.)
| | - Velia D’Agata
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (V.D.)
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Agata Grazia D’Amico
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy;
| |
Collapse
|
6
|
Toledo-Guardiola SM, Luongo C, Abril-Parreño L, Soriano-Úbeda C, Matás C. Different seminal ejaculated fractions in artificial insemination condition the protein cargo of oviductal and uterine extracellular vesicles in pig. Front Cell Dev Biol 2023; 11:1231755. [PMID: 37868907 PMCID: PMC10587466 DOI: 10.3389/fcell.2023.1231755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/21/2023] [Indexed: 10/24/2023] Open
Abstract
The seminal plasma (SP) is the liquid component of semen that facilitates sperm transport through the female genital tract. SP modulates the activity of the ovary, oviductal environment and uterine function during the periovulatory and early pregnancy period. Extracellular vesicles (EVs) secreted in the oviduct (oEVs) and uterus (uEVs) have been shown to influence the expression of endometrial genes that regulate fertilization and early embryo development. In some species, semen is composed of well-separated fractions that vary in concentration of spermatozoa and SP composition and volume. This study aimed to investigate the impact of different accumulative fractions of the porcine ejaculate (F1, composed of the sperm-rich fraction, SRF; F2, composed of F1 plus the intermediate fraction; F3, composed of F2 plus the post-SRF) on oEVs and uEVs protein cargo. Six days after the onset of estrus, we determined the oEVs and uEVs size and protein concentration in pregnant sows by artificial insemination (AI-sows) and in non-inseminated sows as control (C-sows). We also identified the main proteins in oEVs and uEVs, in AI-F1, AI-F2, AI-F3, and C-sows. Our results indicated that although the size of EVs is similar between AI- and C-sows, the protein concentration of both oEVs and uEVs was significantly lower in AI-sows (p < 0.05). Proteomic analysis identified 38 unique proteins in oEVs from AI-sows, mainly involved in protein stabilization, glycolytic and carbohydrate processes. The uEVs from AI-sows showed the presence of 43 unique proteins, including already-known fertility-related proteins (EZR, HSPAA901, PDS). We also demonstrated that the protein composition of oEVs and uEVs differed depending on the seminal fraction(s) inseminated (F1, F2, or F3). In conclusion, we found specific protein cargo in oEVs and uEVs according to the type of semen fraction the sow was inseminated with and whose functions these specific EVs proteins are closely associated with reproductive processes.
Collapse
Affiliation(s)
- S. M. Toledo-Guardiola
- Departamento de Fisiología, Facultad de Veterinaria, Campus de Excelencia Mare Nostrum Universidad de Murcia, Murcia, Spain
| | - C. Luongo
- Departamento de Fisiología, Facultad de Veterinaria, Campus de Excelencia Mare Nostrum Universidad de Murcia, Murcia, Spain
| | - L. Abril-Parreño
- Departamento de Fisiología, Facultad de Veterinaria, Campus de Excelencia Mare Nostrum Universidad de Murcia, Murcia, Spain
| | - C. Soriano-Úbeda
- Departamento de Medicina, Cirugía y Anatomía Veterinaria, Universidad de Léon, León, Spain
| | - C. Matás
- Departamento de Fisiología, Facultad de Veterinaria, Campus de Excelencia Mare Nostrum Universidad de Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB-Arrixaca), Murcia, Spain
| |
Collapse
|
7
|
Zimbone S, Di Rosa MC, Chiechio S, Giuffrida ML. Exploring the Role of Hsp60 in Alzheimer's Disease and Type 2 Diabetes: Suggestion for Common Drug Targeting. Int J Mol Sci 2023; 24:12456. [PMID: 37569831 PMCID: PMC10419248 DOI: 10.3390/ijms241512456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Heat shock protein 60 (Hsp60) is a member of the chaperonin family of heat shock proteins (HSPs), primarily found in the mitochondrial matrix. As a molecular chaperone, Hsp60 plays an essential role in mediating protein folding and assembly, and together with the co-chaperon Hsp10, it is thought to maintain protein homeostasis. Recently, it has been found to localize in non-canonical, extra-mitochondrial sites such as cell membranes or extracellular fluids, particularly in pathological conditions. Starting from its biological function, this review aims to provide a comprehensive understanding of the potential involvement of Hsp60 in Alzheimer's disease (AD) and Type II Diabetes Mellitus (T2DM), which are known to share impaired key pathways and molecular dysfunctions. Fragmentary data reported in the literature reveal interesting links between the altered expression level or localization of this chaperonin and several disease conditions. The present work offers an overview of the past and more recent knowledge about Hsp60 and its role in the most important cellular processes to shed light on neuronal Hsp60 as a potential common target for both pathologies. The absence of any effective cure for AD patients makes the identification of a new molecular target a promising path by which to move forward in the development of new drugs and/or repositioning of therapies already used for T2DM.
Collapse
Affiliation(s)
- Stefania Zimbone
- Institute of Crystallography, National Research Council (CNR-IC), 95126 Catania, Italy; (S.Z.); (M.C.D.R.)
| | - Maria Carmela Di Rosa
- Institute of Crystallography, National Research Council (CNR-IC), 95126 Catania, Italy; (S.Z.); (M.C.D.R.)
- Cogentech Società Benefit srl Actual Position, 95121 Catania, Italy
| | - Santina Chiechio
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy;
- Oasi Research Institute—IRCCS, 94018 Troina, Italy
| | - Maria Laura Giuffrida
- Institute of Crystallography, National Research Council (CNR-IC), 95126 Catania, Italy; (S.Z.); (M.C.D.R.)
| |
Collapse
|
8
|
Paladino L, Santonocito R, Graceffa G, Cipolla C, Pitruzzella A, Cabibi D, Cappello F, Conway de Macario E, Macario AJL, Bucchieri F, Rappa F. Immunomorphological Patterns of Chaperone System Components in Rare Thyroid Tumors with Promise as Biomarkers for Differential Diagnosis and Providing Clues on Molecular Mechanisms of Carcinogenesis. Cancers (Basel) 2023; 15:2403. [PMID: 37190332 PMCID: PMC10136750 DOI: 10.3390/cancers15082403] [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: 03/28/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Hurthle cell (HC), anaplastic (AC), and medullary (MC) carcinomas are low frequency thyroid tumors that pose several challenges for physicians and pathologists due to the scarcity of cases, information, and histopathological images, especially in the many areas around the world in which sophisticated molecular and genetic diagnostic facilities are unavailable. It is, therefore, cogent to provide tools for microscopists to achieve accurate diagnosis, such as histopathological images with reliable biomarkers, which can help them to reach a differential diagnosis. We are investigating whether components of the chaperone system (CS), such as the molecular chaperones, can be considered dependable biomarkers, whose levels and distribution inside and outside cells in the tumor tissue could present a distinctive histopathological pattern for each tumor type. Here, we report data on the chaperones Hsp27, Hsp60, and Hsp90. They presented quantitative levels and distribution patterns that were different for each tumor and differed from those of a benign thyroid pathology, goiter (BG). Therefore, the reported methodology can be beneficial when the microscopist must differentiate between HC, AC, MC, and BG.
Collapse
Affiliation(s)
- Letizia Paladino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy; (L.P.); (R.S.); (A.P.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
| | - Radha Santonocito
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy; (L.P.); (R.S.); (A.P.); (F.C.)
| | - Giuseppa Graceffa
- Department of Surgical Oncology and Oral Sciences, University of Palermo, 90127 Palermo, Italy; (G.G.); (C.C.)
| | - Calogero Cipolla
- Department of Surgical Oncology and Oral Sciences, University of Palermo, 90127 Palermo, Italy; (G.G.); (C.C.)
| | - Alessandro Pitruzzella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy; (L.P.); (R.S.); (A.P.); (F.C.)
- Consortium of Caltanissetta, University of Palermo, 93100 Caltanissetta, Italy
| | - Daniela Cabibi
- Department of Sciences for the Promotion of Health and Mother and Child Care, “G. D’Alessandro”, Pathology Institute, University of Palermo, 90127 Palermo, Italy;
| | - Francesco Cappello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy; (L.P.); (R.S.); (A.P.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
| | - Everly Conway de Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
| | - Alberto J. L. Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy; (L.P.); (R.S.); (A.P.); (F.C.)
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy; (L.P.); (R.S.); (A.P.); (F.C.)
| |
Collapse
|
9
|
De Bakshi D, Chen YC, Wuerzberger-Davis SM, Ma M, Waters BJ, Li L, Suzuki A, Miyamoto S. Ectopic CH60 mediates HAPLN1-induced cell survival signaling in multiple myeloma. Life Sci Alliance 2023; 6:e202201636. [PMID: 36625202 PMCID: PMC9748848 DOI: 10.26508/lsa.202201636] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematological malignancy, is generally considered incurable because of the development of drug resistance. We previously reported that hyaluronan and proteoglycan link protein 1 (HAPLN1) produced by stromal cells induces activation of NF-κB, a tumor-supportive transcription factor, and promotes drug resistance in MM cells. However, the identity of the cell surface receptor that detects HAPLN1 and thereby engenders pro-tumorigenic signaling in MM cells remains unknown. Here, we performed an unbiased cell surface biotinylation assay and identified chaperonin 60 (CH60) as the direct binding partner of HAPLN1 on MM cells. Cell surface CH60 specifically interacted with TLR4 to evoke HAPLN1-induced NF-κB signaling, transcription of anti-apoptotic genes, and drug resistance in MM cells. Collectively, our findings identify a cell surface CH60-TLR4 complex as a HAPLN1 receptor and a potential molecular target to overcome drug resistance in MM cells.
Collapse
Affiliation(s)
- Debayan De Bakshi
- Cellular and Molecular Biology Graduate Program, University of Wisconsin, Madison, WI, USA
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI, USA
- Department of Oncology, University of Wisconsin, Madison, WI, USA
| | - Yu-Chia Chen
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI, USA
- Department of Oncology, University of Wisconsin, Madison, WI, USA
| | - Shelly M Wuerzberger-Davis
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI, USA
- Department of Oncology, University of Wisconsin, Madison, WI, USA
| | - Min Ma
- School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Bayley J Waters
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin, Madison, WI, USA
- Department of Chemistry, University of Wisconsin, Madison, WI, USA
| | - Aussie Suzuki
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI, USA
- Department of Oncology, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Shigeki Miyamoto
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI, USA
- Department of Oncology, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
10
|
Hadizadeh N, Bagheri D, Shamsara M, Hamblin MR, Farmany A, Xu M, Liang Z, Razi F, Hashemi E. Extracellular vesicles biogenesis, isolation, manipulation and genetic engineering for potential in vitro and in vivo therapeutics: An overview. Front Bioeng Biotechnol 2022; 10:1019821. [PMID: 36406206 PMCID: PMC9672340 DOI: 10.3389/fbioe.2022.1019821] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/18/2022] [Indexed: 08/16/2023] Open
Abstract
The main goals of medicine consist of early detection and effective treatment of different diseases. In this regard, the rise of exosomes as carriers of natural biomarkers has recently attracted a lot of attention and managed to shed more light on the future of early disease diagnosis methods. Here, exosome biogenesis, its role as a biomarker in metabolic disorders, and recent advances in state-of-art technologies for exosome detection and isolation will be reviewed along with future research directions and challenges regarding the manipulation and genetic engineering of exosomes for potential in vitro and in vivo disease diagnosis approaches.
Collapse
Affiliation(s)
- Nastaran Hadizadeh
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Diba Bagheri
- Department of Molecular Genetics, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Shamsara
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Abbas Farmany
- Dental Research Centre and Dental Implant Research Centre, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mengdi Xu
- Shenzhen Bay Laboratory, Institute of Molecular Physiology, Shenzhen, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Zhuobin Liang
- Shenzhen Bay Laboratory, Institute of Molecular Physiology, Shenzhen, China
| | - Farideh Razi
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular—Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Hashemi
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- Shenzhen Bay Laboratory, Institute of Molecular Physiology, Shenzhen, China
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular—Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Sun X, Wang X, Yan C, Zheng S, Gao R, Huang F, Wei Y, Wen Z, Chen Y, Zhou X, Liu X, Chen B, Shen Y, Cai Y, Pan N, Wang L. Tumor cell-released LC3-positive EVs promote lung metastasis of breast cancer through enhancing premetastatic niche formation. Cancer Sci 2022; 113:3405-3416. [PMID: 35879596 PMCID: PMC9530874 DOI: 10.1111/cas.15507] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/07/2022] [Accepted: 07/14/2022] [Indexed: 12/24/2022] Open
Abstract
Most breast cancer–related deaths are caused by metastasis in vital organs including the lungs. Development of supportive metastatic microenvironments, referred to as premetastatic niches (PMNs), in certain distant organs before arrival of metastatic cells, is critical in metastasis. However, the mechanisms of PMN formation are not fully clear. Here, we demonstrated that chemoattractant C–C motif chemokine ligand 2 (CCL2) could be stimulated by heat shock protein 60 (HSP60) on the surface of murine 4 T1 breast cancer cell–released LC3+ extracellular vesicles (LC3+ EVs) via the TLR2‐MyD88‐NF‐κB signal cascade in lung fibroblasts, which subsequently promoted lung PMN formation through recruiting monocytes and suppressing T cell function. Consistently, reduction of LC3+ EV release or HSP60 level or neutralization of CCL2 markedly attenuated PMN formation and lung metastasis. Furthermore, the number of circulating LC3+ EVs and HSP60 level on LC3+ EVs in the plasma of breast cancer patients were positively correlated with disease progression and lung metastasis, which might have potential value as biomarkers of lung metastasis in breast cancer patients (AUC = 0.898, 0.694, respectively). These findings illuminate a novel mechanism of PMN formation and might provide therapeutic targets for anti‐metastasis therapy for patients with breast cancer.
Collapse
Affiliation(s)
- Xiaotong Sun
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Xuru Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Chunguang Yan
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Shiya Zheng
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Rong Gao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Fang Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Yiting Wei
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Zhifa Wen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Yongqiang Chen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Xiaohe Zhou
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Xueming Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Bohao Chen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Yuqing Shen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Yunlang Cai
- Department of Obstetrics and Gynecology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Ning Pan
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| | - Lixin Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Immunology, Medical School of Southeast University, Nanjing, China
| |
Collapse
|
12
|
Inigo JR, Chandra D. The mitochondrial unfolded protein response (UPR mt): shielding against toxicity to mitochondria in cancer. J Hematol Oncol 2022; 15:98. [PMID: 35864539 PMCID: PMC9306209 DOI: 10.1186/s13045-022-01317-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/11/2022] [Indexed: 12/20/2022] Open
Abstract
Mitochondria are essential for tumor growth and progression. However, the heavy demand for mitochondrial activity in cancer leads to increased production of mitochondrial reactive oxygen species (mtROS), accumulation of mutations in mitochondrial DNA, and development of mitochondrial dysfunction. If left unchecked, excessive mtROS can damage and unfold proteins in the mitochondria to an extent that becomes lethal to the tumor. Cellular systems have evolved to combat mtROS and alleviate mitochondrial stress through a quality control mechanism called the mitochondrial unfolded protein response (UPRmt). The UPRmt system is composed of chaperones and proteases, which promote protein folding or eliminate mitochondrial proteins damaged by mtROS, respectively. UPRmt is conserved and activated in cancer in response to mitochondrial stress to maintain mitochondrial integrity and support tumor growth. In this review, we discuss how mitochondria become dysfunctional in cancer and highlight the tumor-promoting functions of key components of the UPRmt.
Collapse
Affiliation(s)
- Joseph R Inigo
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
13
|
Wu T, Jia L, Lei S, Jiang H, Liu J, Li N, Langford PR, Liu H, Lei L. Host HSPD1 Translocation from Mitochondria to the Cytoplasm Induced by Streptococcus suis Serovar 2 Enolase Mediates Apoptosis and Loss of Blood-Brain Barrier Integrity. Cells 2022; 11:2071. [PMID: 35805155 PMCID: PMC9265368 DOI: 10.3390/cells11132071] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/01/2023] Open
Abstract
Streptococcus suis serovar 2 (S. suis serovar 2) is a zoonotic pathogen that causes meningitis in pigs and humans, and is a serious threat to the swine industry and public health. Understanding the mechanism(s) by which S. suis serovar 2 penetrates the blood-brain barrier (BBB) is crucial to elucidating the pathogenesis of meningitis. In a previous study, we found that expression of the virulence factor enolase (Eno) by S. suis serovar 2 promotes the expression of host heat shock protein family D member 1 (HSPD1) in brain tissue, which leads to the apoptosis of porcine brain microvascular endothelial cells (PBMECs) and increased BBB permeability, which in turn promotes bacterial translocation across the BBB. However, the mechanism by which HSPD1 mediates Eno-induced apoptosis remains unclear. In this study, we demonstrate that Eno promotes the translocation of HSPD1 from mitochondria to the cytoplasm, where HSPD1 binds to β-actin (ACTB), the translocated HSPD1, and its interaction with ACTB led to adverse changes in cell morphology and promoted the expression of apoptosis-related proteins, second mitochondria-derived activator of caspases (Smac), and cleaved caspase-3; inhibited the expression of X-linked inhibitor of apoptosis protein (XIAP); and finally promoted cell apoptosis. These results further elucidate the role of HSPD1 in the process of Eno-induced apoptosis and increased BBB permeability, increasing our understanding of the pathogenic mechanisms of meningitis, and providing a framework for novel therapeutic strategies.
Collapse
Affiliation(s)
- Tong Wu
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Li Jia
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Siyu Lei
- School of Basic Medicine, Jilin University, Changchun 130021, China;
| | - Hexiang Jiang
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Jianan Liu
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Na Li
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Paul R. Langford
- Section of Paediatric Infectious Disease, Imperial College London, London W2 1NY, UK;
| | - Hongtao Liu
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Liancheng Lei
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
- Department of Veterinary Medicine, College of Animal Science, Yangtze University, Jingzhou 434023, China
| |
Collapse
|
14
|
Tang Y, Zhou Y, Fan S, Wen Q. The Multiple Roles and Therapeutic Potential of HSP60 in Cancer. Biochem Pharmacol 2022; 201:115096. [DOI: 10.1016/j.bcp.2022.115096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023]
|
15
|
Rathi D, Verma JK, Pareek A, Chakraborty S, Chakraborty N. Dissection of grasspea (Lathyrus sativus L.) root exoproteome reveals critical insights and novel proteins. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2022; 316:111161. [PMID: 35151446 DOI: 10.1016/j.plantsci.2021.111161] [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: 07/09/2021] [Revised: 11/20/2021] [Accepted: 12/18/2021] [Indexed: 06/14/2023]
Abstract
The plant exoproteome is crucial because its constituents greatly influence plant phenotype by regulating physiological characteristics to adapt to environmental stresses. The root exudates constitute a dynamic aspect of plant exoproteome, as its molecular composition ensures a beneficial rhizosphere in a species-specific manner. We investigated the root exoproteome of grasspea, a stress-resilient pulse and identified 2861 non-redundant proteins, belonging to a myriad of functional classes, including root development, rhizosphere augmentation as well as defense functions against soil-borne pathogens. Significantly, we identified 1986 novel exoproteome constituents of grasspea, potentially involved in cell-to-cell communication and root meristem maintenance, among other critical roles. Sequence-based comparison revealed that grasspea shares less than 30 % of its exoproteome with the reports so far from model plants as well as crop species. Further, the exoproteome revealed 65 % proteins to be extracellular in nature and of these, 37 % constituents were predicted to follow unconventional protein secretion (UPS) mode. We validated the UPS for four stress-responsive proteins, which were otherwise predicted to follow classical protein secretion (CPS). Conclusively, we recognized not only the highest number of root exudate proteins, but also pinpointed novel signatures of dicot root exoproteome.
Collapse
Affiliation(s)
- Divya Rathi
- National Institute of Plant Genome Research, Jawaharlal Nehru University Campus, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Jitendra Kumar Verma
- National Institute of Plant Genome Research, Jawaharlal Nehru University Campus, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Akanksha Pareek
- National Institute of Plant Genome Research, Jawaharlal Nehru University Campus, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Subhra Chakraborty
- National Institute of Plant Genome Research, Jawaharlal Nehru University Campus, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Niranjan Chakraborty
- National Institute of Plant Genome Research, Jawaharlal Nehru University Campus, Aruna Asaf Ali Marg, New Delhi, 110067, India.
| |
Collapse
|
16
|
Exosomes and Other Extracellular Vesicles with High Therapeutic Potential: Their Applications in Oncology, Neurology, and Dermatology. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041303. [PMID: 35209095 PMCID: PMC8879284 DOI: 10.3390/molecules27041303] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023]
Abstract
Until thirty years ago, it was believed that extracellular vesicles (EVs) were used to remove unnecessary compounds from the cell. Today, we know about their enormous potential in diagnosing and treating various diseases. EVs are essential mediators of intercellular communication, enabling the functional transfer of bioactive molecules from one cell to another. Compared to laboratory-created drug nanocarriers, they are stable in physiological conditions. Furthermore, they are less immunogenic and cytotoxic compared to polymerized vectors. Finally, EVs can transfer cargo to particular cells due to their membrane proteins and lipids, which can implement them to specific receptors in the target cells. Recently, new strategies to produce ad hoc exosomes have been devised. Cells delivering exosomes have been genetically engineered to overexpress particular macromolecules, or transformed to release exosomes with appropriate targeting molecules. In this way, we can say tailor-made therapeutic EVs are created. Nevertheless, there are significant difficulties to solve during the application of EVs as drug-delivery agents in the clinic. This review explores the diversity of EVs and the potential therapeutic options for exosomes as natural drug-delivery vehicles in oncology, neurology, and dermatology. It also reflects future challenges in clinical translation.
Collapse
|
17
|
Correll VL, Otto JJ, Risi CM, Main BP, Boutros PC, Kislinger T, Galkin VE, Nyalwidhe JO, Semmes OJ, Yang L. Optimization of small extracellular vesicle isolation from expressed prostatic secretions in urine for in-depth proteomic analysis. J Extracell Vesicles 2022; 11:e12184. [PMID: 35119778 PMCID: PMC8815402 DOI: 10.1002/jev2.12184] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/22/2021] [Accepted: 12/22/2021] [Indexed: 01/23/2023] Open
Abstract
The isolation and subsequent molecular analysis of extracellular vesicles (EVs) derived from patient samples is a widely used strategy to understand vesicle biology and to facilitate biomarker discovery. Expressed prostatic secretions in urine are a tumor proximal fluid that has received significant attention as a source of potential prostate cancer (PCa) biomarkers for use in liquid biopsy protocols. Standard EV isolation methods like differential ultracentrifugation (dUC) co-isolate protein contaminants that mask lower-abundance proteins in typical mass spectrometry (MS) protocols. Further complicating the analysis of expressed prostatic secretions, uromodulin, also known as Tamm-Horsfall protein (THP), is present at high concentrations in urine. THP can form polymers that entrap EVs during purification, reducing yield. Disruption of THP polymer networks with dithiothreitol (DTT) can release trapped EVs, but smaller THP fibres co-isolate with EVs during subsequent ultracentrifugation. To resolve these challenges, we describe here a dUC method that incorporates THP polymer reduction and alkaline washing to improve EV isolation and deplete both THP and other common protein contaminants. When applied to human expressed prostatic secretions in urine, we achieved relative enrichment of known prostate and prostate cancer-associated EV-resident proteins. Our approach provides a promising strategy for global proteomic analyses of urinary EVs.
Collapse
Affiliation(s)
- Vanessa L. Correll
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Joseph J. Otto
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Cristina M. Risi
- Department of Physiological SciencesEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Brian P. Main
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Paul C. Boutros
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoCanada
- Department of Human GeneticsUniversity of CaliforniaLos AngelesCaliforniaUSA
- Department of UrologyUniversity of CaliforniaLos AngelesCaliforniaUSA
- Institute for Precision HealthUniversity of CaliforniaLos AngelesCaliforniaUSA
- Jonsson Comprehensive Cancer CenterUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Thomas Kislinger
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
| | - Vitold E. Galkin
- Department of Physiological SciencesEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Julius O. Nyalwidhe
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - O. John Semmes
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Lifang Yang
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
| |
Collapse
|
18
|
Cancer extracellular vesicles, tumoroid models, and tumor microenvironment. Semin Cancer Biol 2022; 86:112-126. [PMID: 35032650 DOI: 10.1016/j.semcancer.2022.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/21/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022]
Abstract
Cancer extracellular vesicles (EVs), or exosomes, promote tumor progression through enhancing tumor growth, initiating epithelial-to-mesenchymal transition, remodeling the tumor microenvironment, and preparing metastatic niches. Three-dimensionally (3D) cultured tumoroids / spheroids aim to reproduce some aspects of tumor behavior in vitro and show increased cancer stem cell properties. These properties are transferred to their EVs that promote tumor growth. Moreover, recent tumoroid models can be furnished with aspects of the tumor microenvironment, such as vasculature, hypoxia, and extracellular matrix. This review summarizes tumor tissue culture and engineering platforms compatible with EV research. For example, the combination experiments of 3D-tumoroids and EVs have revealed multifunctional proteins loaded in EVs, such as metalloproteinases and heat shock proteins. EVs or exosomes are able to transfer their cargo molecules to recipient cells, whose fates are often largely altered. In addition, the review summarizes approaches to EV labeling technology using fluorescence and luciferase, useful for studies on EV-mediated intercellular communication, biodistribution, and metastatic niche formation.
Collapse
|
19
|
Abu N, Rus Bakarurraini NAA, Nasir SN. Extracellular Vesicles and DAMPs in Cancer: A Mini-Review. Front Immunol 2021; 12:740548. [PMID: 34721407 PMCID: PMC8554306 DOI: 10.3389/fimmu.2021.740548] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/07/2021] [Indexed: 12/24/2022] Open
Abstract
Certain cancer therapy has been shown to induce immunogenic cell death in cancer cells and may promote tumor progression instead. The external stress or stimuli may induce cell death and contribute toward the secretion of pro inflammatory molecules. The release of damage-associated molecular patterns (DAMPs) upon induction of therapy or cell death has been shown to induce an inflammatory response. Nevertheless, the mechanism as to how the DAMPs are released and engage in such activity needs further in-depth investigation. Interestingly, some studies have shown that DAMPs can be released through extracellular vesicles (EVs) and can bind to receptors such as toll-like receptors (TCRs). Ample pre-clinical studies have shown that cancer-derived EVs are able to modulate immune responses within the tumor microenvironment. However, the information on the presence of such DAMPs within EVs is still elusive. Therefore, this mini-review attempts to summarize and appraise studies that have shown the presence of DAMPs within cancer-EVs and how it affects the downstream cellular process.
Collapse
Affiliation(s)
- Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | | | - Siti Nurmi Nasir
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
20
|
Caruso Bavisotto C, Marino Gammazza A, Campanella C, Bucchieri F, Cappello F. Extracellular heat shock proteins in cancer: From early diagnosis to new therapeutic approach. Semin Cancer Biol 2021; 86:36-45. [PMID: 34563652 DOI: 10.1016/j.semcancer.2021.09.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/10/2021] [Accepted: 09/19/2021] [Indexed: 02/08/2023]
Abstract
In cancer, human cells lose the ability to properly control the series of events that occur constantly during cell growth and division, including protein expression, stability, and dynamics. Heat shock proteins (Hsps) are key molecules in these events, constitutively expressed at high levels and could furthermore be induced by the response to cancer-induced stress. In tumor cells, Hsps have been shown to be implicated in the regulation of apoptosis, immune responses, angiogenesis and metastasis; in some cases, they can be overexpressed and dysregulated, representing important cancer hallmarks. In the past few years, it has been demonstrated that Hsps can be released by tumor cells through several secreting pathways, including the extracellular vesicles (EVs), thus modulating the tumor microenvironment as well as long-distance intercellular communication and metastatization. In this review, we discuss the role of extracellular Hsps in cancer, with a particular interest in Hsps in EVs. We would also like to highlight the importance of fully understanding of the role of extracellular Hsps released by EVs and encourage further research in this field the use of Hsps as early cancer biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Celeste Caruso Bavisotto
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Anatomy and Histology, University of Palermo, Palermo, Italy; Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Claudia Campanella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Fabio Bucchieri
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Francesco Cappello
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Anatomy and Histology, University of Palermo, Palermo, Italy; Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy.
| |
Collapse
|
21
|
Regimbeau M, Abrey J, Vautrot V, Causse S, Gobbo J, Garrido C. Heat shock proteins and exosomes in cancer theranostics. Semin Cancer Biol 2021; 86:46-57. [PMID: 34343652 DOI: 10.1016/j.semcancer.2021.07.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 01/19/2023]
Abstract
Heat shock proteins (HSPs) are a superfamily of molecular chaperones that were discovered through their ability to be induced by different stresses including heat shock. Other than their function as chaperones in proteins homeostasis, HSPs have been shown to inhibit different forms of cell death and to participate in cell proliferation and differentiation processes. Because cancer cells have to rewire their metabolism, they require a high amount of these stress-inducible chaperones for their survival. Therefore, HSPs are unusually abundant in cancer cells where they have oncogene-like functions. In cancer, HSPs have been involved in the regulation of apoptosis, immune responses, angiogenesis, metastasis and treatment resistance. Recently, HSPs have been shown to be secreted through exosomes by cancer cells. These tumor-derived exosomes can be used as circulating markers: HSP-exosomes have been reported as biomarkers of cancer dissemination, response to therapy and/or patient outcome. A new range of functions, mostly in modulation of anticancer immune responses, have been described for these extracellular HSPs. In this review, we will describe those recently reported functions of HSP-exosomes that makes them both targets for anticancer therapeutics and biomarkers for the monitoring of the disease. We will also discuss their emerging interest in cancer vaccines.
Collapse
Affiliation(s)
- Mathilde Regimbeau
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Université. Bourgogne Franche-Comté, 21000, Dijon, France
| | - Jimena Abrey
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Université. Bourgogne Franche-Comté, 21000, Dijon, France
| | - Valentin Vautrot
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Université. Bourgogne Franche-Comté, 21000, Dijon, France; Anticancer Center Georges François Leclerc, Dijon, France
| | - Sebastien Causse
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Université. Bourgogne Franche-Comté, 21000, Dijon, France
| | - Jessica Gobbo
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Anticancer Center Georges François Leclerc, Dijon, France; Early Phase Unit INCa CLIP², Department of Oncology, Georges-François Leclerc Centre, Dijon, France; Centre d'investigation Clinique INSERM 1432, CHU Dijon-Bourgogne, Dijon, France
| | - Carmen Garrido
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Université. Bourgogne Franche-Comté, 21000, Dijon, France; Anticancer Center Georges François Leclerc, Dijon, France.
| |
Collapse
|
22
|
Singh P, Kumari M, Bal A, Srinivasan R, Ghosh S. Heat shock protein 60 is a disease-associated sialoglycoprotein in human non-small cell lung cancer. Biol Chem 2021; 401:969-983. [PMID: 32049642 DOI: 10.1515/hsz-2019-0352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 02/03/2020] [Indexed: 01/23/2023]
Abstract
The diagnostic and therapeutic potential of Maackia amurensis agglutinin (MAA) have been reported in various malignancies. Earlier, we have found that MAA specifically interacted with human non-small cell lung-cancer (NSCLC) cells and induced apoptosis in these cells. The present study was designed to identify M. amurensis leukoagglutinin (MAL-I, one of the components of MAA, having the same carbohydrate specificity as MAA) interacting membrane sialoglycoprotein(s) of two subtypes of human NSCLC cell lines. Nine proteins were identified using two-dimensional (2D)-polyacrylamide gel electrophoresis (PAGE) followed by MAL-I-overlay transblotting and matrix assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF-MS). Among these proteins, HSP60 was selected for further characterization. The sialoglycoprotein nature of membrane-HSP60 of NSCLC cell lines was confirmed by its reduced reactivity with MAL-I in Western blots in the presence of GM2 and by dual staining of the cell lines with MAL-I and HSP60-antibody. These findings were further substantiated by enzymatic analysis of membrane-HSP60 as well as in-silico evidence regarding this protein. Our observations were validated by immunohistochemical analysis of both subtypes of NSCLC tissue sections. Membrane-HSP60 was found to be involved in the inhibition of MAL-I-induced morphological alteration of NSCLC cells and also in the proliferation and migration of these cells, indicating the probable role of sialylated membrane-HSP60 in this disease.
Collapse
Affiliation(s)
- Praveen Singh
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh 160012, India
| | - Munmun Kumari
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh 160012, India
| | - Amanjit Bal
- Department of Histopathology, PGIMER, Chandigarh 160012, India
| | - Radhika Srinivasan
- Department of Cytology and Gynecological Pathology, PGIMER, Chandigarh 160012, India
| | - Sujata Ghosh
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh 160012, India
| |
Collapse
|
23
|
Sun B, Li G, Yu Q, Liu D, Tang X. HSP60 in cancer: a promising biomarker for diagnosis and a potentially useful target for treatment. J Drug Target 2021; 30:31-45. [PMID: 33939586 DOI: 10.1080/1061186x.2021.1920025] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Heat shock proteins (HSPs), most of which are molecular chaperones, are highly conserved proteins produced by cells under physiological stress or pathological conditions. HSP60 (57-69 kDa) can promote or inhibit cell apoptosis through different mechanisms, and its abnormal expression is also related to tumour cell metastasis and drug resistance. In recent years, HSP60 has received increasing attention in the field of cancer research due to its potential as a diagnostic and prognostic biomarker or therapeutic target. However, in different types of cancer, the specific mechanisms of abnormally expressed HSP60 in tumour carcinogenesis and drug resistance are complicated and still require further study. In this article, we comprehensively review the regulative mechanisms of HSP60 on apoptosis, its applications as a cancer diagnostic biomarker and a therapeutic target, evidence of involvement in tumour resistance and the applications of exosomal HSP60 in liquid biopsy. By evaluating the current findings of HSP60 in cancer research, we highlight some core issues that need to be addressed for the use of HSP60 as a diagnostic or prognostic biomarker and therapeutic target in certain types of cancer.
Collapse
Affiliation(s)
- Bo Sun
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Ganghui Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Qing Yu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Dongchun Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Xing Tang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, PR China
| |
Collapse
|
24
|
Molecular Chaperones and Thyroid Cancer. Int J Mol Sci 2021; 22:ijms22084196. [PMID: 33919591 PMCID: PMC8073690 DOI: 10.3390/ijms22084196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/17/2022] Open
Abstract
Thyroid cancers are the most common of the endocrine system malignancies and progress must be made in the areas of differential diagnosis and treatment to improve patient management. Advances in the understanding of carcinogenic mechanisms have occurred in various fronts, including studies of the chaperone system (CS). Components of the CS are found to be quantitatively increased or decreased, and some correlations have been established between the quantitative changes and tumor type, prognosis, and response to treatment. These correlations provide the basis for identifying distinctive patterns useful in differential diagnosis and for planning experiments aiming at elucidating the role of the CS in tumorigenesis. Here, we discuss studies of the CS components in various thyroid cancers (TC). The chaperones belonging to the families of the small heat-shock proteins Hsp70 and Hsp90 and the chaperonin of Group I, Hsp60, have been quantified mostly by immunohistochemistry and Western blot in tumor and normal control tissues and in extracellular vesicles. Distinctive differences were revealed between the various thyroid tumor types. The most frequent finding was an increase in the chaperones, which can be attributed to the augmented need for chaperones the tumor cells have because of their accelerated metabolism, growth, and division rate. Thus, chaperones help the tumor cell rather than protect the patient, exemplifying chaperonopathies by mistake or collaborationism. This highlights the need for research on chaperonotherapy, namely the development of means to eliminate/inhibit pathogenic chaperones.
Collapse
|
25
|
The Triad Hsp60-miRNAs-Extracellular Vesicles in Brain Tumors: Assessing Its Components for Understanding Tumorigenesis and Monitoring Patients. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11062867] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Brain tumors have a poor prognosis and progress must be made for developing efficacious treatments, but for this to occur their biology and interaction with the host must be elucidated beyond current knowledge. What has been learned from other tumors may be applied to study brain tumors, for example, the role of Hsp60, miRNAs, and extracellular vesicles (EVs) in the mechanisms of cell proliferation and dissemination, and resistance to immune attack and anticancer drugs. It has been established that Hsp60 increases in cancer cells, in which it occurs not only in the mitochondria but also in the cytosol and plasma-cell membrane and it is released in EVs into the extracellular space and in circulation. There is evidence suggesting that these EVs interact with cells near and far from their original cell and that this interaction has an impact on the functions of the target cell. It is assumed that this crosstalk between cancer and host cells favors carcinogenesis in various ways. We, therefore, propose to study the triad Hsp60-related miRNAs-EVs in brain tumors and have standardized methods for the purpose. These revealed that EVs with Hsp60 and related miRNAs increase in patients’ blood in a manner that reflects disease status. The means are now available to monitor brain tumor patients by measuring the triad and to dissect its effects on target cells in vitro, and in experimental models in vivo.
Collapse
|
26
|
Schwartz RE, Shokhirev MN, Andrade LR, Gutkind JS, Iglesias-Bartolome R, Shadel GS. Insights into epithelial cell senescence from transcriptome and secretome analysis of human oral keratinocytes. Aging (Albany NY) 2021; 13:4747-4777. [PMID: 33601339 PMCID: PMC7950289 DOI: 10.18632/aging.202658] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/21/2021] [Indexed: 01/08/2023]
Abstract
Senescent cells produce chronic inflammation that contributes to the diseases and debilities of aging. How this process is orchestrated in epithelial cells, the origin of human carcinomas, is poorly understood. We used human normal oral keratinocytes (NOKs) to elucidate senescence programs in a prototype primary mucosal epithelial cell that senesces spontaneously. While NOKs exhibit several typical facets of senescence, they also display distinct characteristics. These include expression of p21WAF1/CIP1 at early passages, making this common marker of senescence unreliable in NOKs. Transcriptome analysis by RNA-seq revealed specific commonalities with and differences from cancer cells, explicating the tumor avoidance role of senescence. Repression of DNA repair genes that correlated with downregulation of E2F1 mRNA and protein was observed for two donors; a divergent result was seen for the third. Using proteomic profiling of soluble (non-vesicular) and extracellular vesicle (EV) associated secretions, we propose additions to the senescence associated secretory phenotype, including HSP60, which localizes to the surface of EVs. Finally, EVs from senescent NOKs activate interferon pathway signaling in THP-1 monocytes in a STING-dependent manner and associate with mitochondrial and nuclear DNA. Our results highlight senescence changes in epithelial cells and how they might contribute to chronic inflammation and age-related diseases.
Collapse
Affiliation(s)
- Rachael E Schwartz
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies and University of California - San Diego, La Jolla, CA 92037, USA
| | - Maxim N Shokhirev
- Razavi Newman Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Leonardo R Andrade
- Waitt Advanced Biophotonics Core, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - J Silvio Gutkind
- Department of Pharmacology and Moores Cancer Center, University of California - San Diego, La Jolla, CA 92093, USA
| | - Ramiro Iglesias-Bartolome
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gerald S Shadel
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies and University of California - San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
27
|
D’Amico D, Fiore R, Caporossi D, Di Felice V, Cappello F, Dimauro I, Barone R. Function and Fiber-Type Specific Distribution of Hsp60 and αB-Crystallin in Skeletal Muscles: Role of Physical Exercise. BIOLOGY 2021; 10:biology10020077. [PMID: 33494467 PMCID: PMC7911561 DOI: 10.3390/biology10020077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/19/2022]
Abstract
Simple Summary Skeletal muscle represents about 40% of the body mass in humans and it is a copious and plastic tissue, rich in proteins that are subject to continuous rearrangements. Physical exercise is considered a physiological stressor for different organs, in particular for skeletal muscle, and it is a factor able to stimulate the cellular remodeling processes related to the phenomenon of adaptation. All cells respond to various stress conditions by up-regulating the expression and/or activation of a group of proteins called heat shock proteins (HSPs). Although their expression is induced by several stimuli, they are commonly recognized as HSPs due to the first experiments showing their increased transcription after application of heat shock. These proteins are molecular chaperones mainly involved in assisting protein transport and folding, assembling multimolecular complexes, and triggering protein degradation by proteasome. Among the HSPs, a special attention needs to be devoted to Hsp60 and αB-crystallin, proteins constitutively expressed in the skeletal muscle, where they are known to be important in muscle physiopathology. Therefore, here we provide a critical update on their role in skeletal muscle fibers after physical exercise, highlighting the control of their expression, their biological function, and their specific distribution within skeletal muscle fiber-types. Abstract Skeletal muscle is a plastic and complex tissue, rich in proteins that are subject to continuous rearrangements. Skeletal muscle homeostasis can be affected by different types of stresses, including physical activity, a physiological stressor able to stimulate a robust increase in different heat shock proteins (HSPs). The modulation of these proteins appears to be fundamental in facilitating the cellular remodeling processes related to the phenomenon of training adaptations such as hypertrophy, increased oxidative capacity, and mitochondrial activity. Among the HSPs, a special attention needs to be devoted to Hsp60 and αB-crystallin (CRYAB), proteins constitutively expressed in the skeletal muscle, where their specific features could be highly relevant in understanding the impact of different volumes of training regimes on myofiber types and in explaining the complex picture of exercise-induced mechanical strain and damaging conditions on fiber population. This knowledge could lead to a better personalization of training protocols with an optimal non-harmful workload in populations of individuals with different needs and healthy status. Here, we introduce for the first time to the reader these peculiar HSPs from the perspective of exercise response, highlighting the control of their expression, biological function, and specific distribution within skeletal muscle fiber-types.
Collapse
Affiliation(s)
- Daniela D’Amico
- Human Anatomy Section, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (D.D.); (V.D.F.)
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, TX 77554, USA
| | - Roberto Fiore
- Postgraduate School of Sports Medicine, University Hospital of Palermo, 90127 Palermo, Italy;
| | - Daniela Caporossi
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy;
| | - Valentina Di Felice
- Human Anatomy Section, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (D.D.); (V.D.F.)
| | - Francesco Cappello
- Human Anatomy Section, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (D.D.); (V.D.F.)
- Euro-Mediterranean Institutes of Science and Technology (IEMEST), 90139 Palermo, Italy
- Correspondence: (F.C.); (I.D.); (R.B.); Tel.: +39-091-2386-5823 (F.C. & R.B.); +39-06-3673-3562 (I.D.)
| | - Ivan Dimauro
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy;
- Correspondence: (F.C.); (I.D.); (R.B.); Tel.: +39-091-2386-5823 (F.C. & R.B.); +39-06-3673-3562 (I.D.)
| | - Rosario Barone
- Human Anatomy Section, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (D.D.); (V.D.F.)
- Correspondence: (F.C.); (I.D.); (R.B.); Tel.: +39-091-2386-5823 (F.C. & R.B.); +39-06-3673-3562 (I.D.)
| |
Collapse
|
28
|
Adamo A, Frusteri C, Pallotta MT, Pirali T, Sartoris S, Ugel S. Moonlighting Proteins Are Important Players in Cancer Immunology. Front Immunol 2021; 11:613069. [PMID: 33584695 PMCID: PMC7873856 DOI: 10.3389/fimmu.2020.613069] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
Plasticity and adaptation to environmental stress are the main features that tumor and immune system share. Except for intrinsic and high-defined properties, cancer and immune cells need to overcome the opponent's defenses by activating more effective signaling networks, based on common elements such as transcriptional factors, protein-based complexes and receptors. Interestingly, growing evidence point to an increasing number of proteins capable of performing diverse and unpredictable functions. These multifunctional proteins are defined as moonlighting proteins. During cancer progression, several moonlighting proteins are involved in promoting an immunosuppressive microenvironment by reprogramming immune cells to support tumor growth and metastatic spread. Conversely, other moonlighting proteins support tumor antigen presentation and lymphocytes activation, leading to several anti-cancer immunological responses. In this light, moonlighting proteins could be used as promising new potential targets for improving current cancer therapies. In this review, we describe in details 12 unprecedented moonlighting proteins that during cancer progression play a decisive role in guiding cancer-associated immunomodulation by shaping innate or adaptive immune response.
Collapse
Affiliation(s)
- Annalisa Adamo
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Cristina Frusteri
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | | | - Tracey Pirali
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Silvia Sartoris
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Stefano Ugel
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
29
|
Functions and Therapeutic Potential of Extracellular Hsp60, Hsp70, and Hsp90 in Neuroinflammatory Disorders. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11020736] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuroinflammation is implicated in central nervous system (CNS) diseases, but the molecular mechanisms involved are poorly understood. Progress may be accelerated by developing a comprehensive view of the pathogenesis of CNS disorders, including the immune and the chaperone systems (IS and CS). The latter consists of the molecular chaperones; cochaperones; and chaperone cofactors, interactors, and receptors of an organism and its main collaborators in maintaining protein homeostasis (canonical function) are the ubiquitin–proteasome system and chaperone-mediated autophagy. The CS has also noncanonical functions, for instance, modulation of the IS with induction of proinflammatory cytokines. This deserves investigation because it may be at the core of neuroinflammation, and elucidation of its mechanism will open roads toward developing efficacious treatments centered on molecular chaperones (i.e., chaperonotherapy). Here, we discuss information available on the role of three members of the CS—heat shock protein (Hsp)60, Hsp70, and Hsp90—in IS modulation and neuroinflammation. These three chaperones occur intra- and extracellularly, with the latter being the most likely involved in neuroinflammation because they can interact with the IS. We discuss some of the interactions, their consequences, and the molecules involved but many aspects are still incompletely elucidated, and we hope that this review will encourage research based on the data presented to pave the way for the development of chaperonotherapy. This may consist of blocking a chaperone that promotes destructive neuroinflammation or replacing or boosting a defective chaperone with cytoprotective activity against neurodegeneration.
Collapse
|
30
|
Krawczyk MA, Pospieszynska A, Styczewska M, Bien E, Sawicki S, Marino Gammazza A, Fucarino A, Gorska-Ponikowska M. Extracellular Chaperones as Novel Biomarkers of Overall Cancer Progression and Efficacy of Anticancer Therapy. APPLIED SCIENCES 2020; 10:6009. [DOI: 10.3390/app10176009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Exosomal heat shock proteins (Hsps) are involved in intercellular communication both in physiological and pathological conditions. They play a role in key processes of carcinogenesis including immune system regulation, cell differentiation, vascular homeostasis and metastasis formation. Thus, exosomal Hsps are emerging biomarkers of malignancies and possible therapeutic targets. Adolescents and young adults (AYAs) are patients aged 15–39 years. This age group, placed between pediatric and adult oncology, pose a particular challenge for cancer management. New biomarkers of cancer growth and progression as well as prognostic factors are desperately needed in AYAs. In this review, we attempted to summarize the current knowledge on the role of exosomal Hsps in selected solid tumors characteristic for the AYA population and/or associated with poor prognosis in this age group. These included malignant melanoma, brain tumors, and breast, colorectal, thyroid, hepatocellular, lung and gynecological tract carcinomas. The studies on exosomal Hsps in these tumors are limited; however; some have provided promising results. Although further research is needed, there is potential for future clinical applications of exosomal Hsps in AYA cancers, both as novel biomarkers of disease presence, progression or relapse, or as therapeutic targets or tools for drug delivery.
Collapse
|
31
|
Extracellular Vesicles-Based Drug Delivery Systems: A New Challenge and the Exemplum of Malignant Pleural Mesothelioma. Int J Mol Sci 2020; 21:ijms21155432. [PMID: 32751556 PMCID: PMC7432055 DOI: 10.3390/ijms21155432] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
Research for the most selective drug delivery to tumors represents a fascinating key target in science. Alongside the artificial delivery systems identified in the last decades (e.g., liposomes), a family of natural extracellular vesicles (EVs) has gained increasing focus for their potential use in delivering anticancer compounds. EVs are released by all cell types to mediate cell-to-cell communication both at the paracrine and the systemic levels, suggesting a role for them as an ideal nano-delivery system. Malignant pleural mesothelioma (MPM) stands out among currently untreatable tumors, also due to the difficulties in achieving an early diagnosis. Thus, early diagnosis and treatment of MPM are both unmet clinical needs. This review looks at indirect and direct evidence that EVs may represent both a new tool for allowing an early diagnosis of MPM and a potential new delivery system for more efficient therapeutic strategies. Since MPM is a relatively rare malignant tumor and preclinical MPM models developed to date are very few and not reliable, this review will report data obtained in other tumor types, suggesting the potential use of EVs in mesothelioma patients as well.
Collapse
|
32
|
Rodriguez A, Von Salzen D, Holguin BA, Bernal RA. Complex Destabilization in the Mitochondrial Chaperonin Hsp60 Leads to Disease. Front Mol Biosci 2020; 7:159. [PMID: 32766281 PMCID: PMC7381220 DOI: 10.3389/fmolb.2020.00159] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/24/2020] [Indexed: 01/21/2023] Open
Abstract
Several neurological disorders have been linked to mutations in chaperonin genes and more specifically to the HSPD1 gene. In humans, HSPD1 encodes the mitochondrial Heat Shock Protein 60 (mtHsp60) chaperonin, which carries out essential protein folding reactions that help maintain mitochondrial and cellular homeostasis. It functions as a macromolecular complex that provides client proteins an environment that favors proper folding in an ATP-dependent manner. It has been established that mtHsp60 plays a crucial role in the proper folding of mitochondrial proteins involved in ATP producing pathways. Recently, various single-point mutations in the mtHsp60 encoding gene have been directly linked to neuropathies and paraplegias. Individuals who harbor mtHsp60 mutations that negatively impact its folding ability display phenotypes with highly compromised muscle and neuron cells. Carriers of these mutations usually develop neuropathies and paraplegias at different stages of their lives mainly characterized by leg stiffness and weakness as well as degeneration of spinal cord nerves. These phenotypes are likely due to hindered energy producing pathways involved in cellular respiration resulting in ATP deprived cells. Although the complete protein folding mechanism of mtHsp60 is not well understood, recent work suggests that several of these mutations act by destabilizing the oligomeric stability of mtHsp60. Here, we discuss recent studies that highlight key aspects of the mtHsp60 mechanism with a focus on some of the known disease-causing point mutations, D29G and V98I, and their effect on the protein folding reaction cycle.
Collapse
Affiliation(s)
| | | | | | - Ricardo A. Bernal
- Department of Chemistry and Biochemistry, The University of Texas at El Paso, El Paso, TX, United States
| |
Collapse
|
33
|
Cappello F, Marino Gammazza A, Dieli F, Conway de Macario E, Macario AJL. Does SARS-CoV-2 Trigger Stress-InducedAutoimmunity by Molecular Mimicry? A Hypothesis. J Clin Med 2020; 9:jcm9072038. [PMID: 32610587 PMCID: PMC7408943 DOI: 10.3390/jcm9072038] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/11/2020] [Accepted: 06/26/2020] [Indexed: 01/08/2023] Open
Abstract
Viruses can generate molecular mimicry phenomena within their hosts. Why should severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) not be considered one of these? Information in this short review suggests that it might be so and, thus, encourages research aiming at testing this possibility. We propose, as a working hypothesis, that the virus induces antibodies and that some of them crossreact with host’s antigens, thus eliciting autoimmune phenomena with devasting consequences in various tissues and organs. If confirmed, by in vitro and in vivo tests, this could drive researchers to find effective treatments against the virus.
Collapse
Affiliation(s)
- Francesco Cappello
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.G.); (F.D.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90141 Palermo, Italy;
- Correspondence: (F.C.); (A.J.L.M.)
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.G.); (F.D.)
| | - Francesco Dieli
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.G.); (F.D.)
| | - Everly Conway de Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90141 Palermo, Italy;
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
| | - Alberto JL Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90141 Palermo, Italy;
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
- Correspondence: (F.C.); (A.J.L.M.)
| |
Collapse
|
34
|
Caruso Bavisotto C, Alberti G, Vitale AM, Paladino L, Campanella C, Rappa F, Gorska M, Conway de Macario E, Cappello F, Macario AJL, Marino Gammazza A. Hsp60 Post-translational Modifications: Functional and Pathological Consequences. Front Mol Biosci 2020; 7:95. [PMID: 32582761 PMCID: PMC7289027 DOI: 10.3389/fmolb.2020.00095] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022] Open
Abstract
Hsp60 is a chaperone belonging to the Chaperonins of Group I and typically functions inside mitochondria in which, together with the co-chaperonin Hsp10, maintains protein homeostasis. In addition to this canonical role, Hsp60 plays many others beyond the mitochondria, for instance in the cytosol, plasma-cell membrane, extracellular space, and body fluids. These non-canonical functions include participation in inflammation, autoimmunity, carcinogenesis, cell replication, and other cellular events in health and disease. Thus, Hsp60 is a multifaceted molecule with a wide range of cellular and tissue locations and functions, which is noteworthy because there is only one hsp60 gene. The question is by what mechanism this protein can become multifaceted. Likely, one factor contributing to this diversity is post-translational modification (PTM). The amino acid sequence of Hsp60 contains many potential phosphorylation sites, and other PTMs are possible such as O-GlcNAcylation, nitration, acetylation, S-nitrosylation, citrullination, oxidation, and ubiquitination. The effect of some of these PTMs on Hsp60 functions have been examined, for instance phosphorylation has been implicated in sperm capacitation, docking of H2B and microtubule-associated proteins, mitochondrial dysfunction, tumor invasiveness, and delay or facilitation of apoptosis. Nitration was found to affect the stability of the mitochondrial permeability transition pore, to inhibit folding ability, and to perturb insulin secretion. Hyperacetylation was associated with mitochondrial failure; S-nitrosylation has an impact on mitochondrial stability and endothelial integrity; citrullination can be pro-apoptotic; oxidation has a role in the response to cellular injury and in cell migration; and ubiquitination regulates interaction with the ubiquitin-proteasome system. Future research ought to determine which PTM causes which variations in the Hsp60 molecular properties and functions, and which of them are pathogenic, causing chaperonopathies. This is an important topic considering the number of acquired Hsp60 chaperonopathies already cataloged, many of which are serious diseases without efficacious treatment.
Collapse
Affiliation(s)
- Celeste Caruso Bavisotto
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostic (BIND), University of Palermo, Palermo, Italy.,Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | - Giusi Alberti
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostic (BIND), University of Palermo, Palermo, Italy
| | - Alessandra Maria Vitale
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostic (BIND), University of Palermo, Palermo, Italy
| | - Letizia Paladino
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostic (BIND), University of Palermo, Palermo, Italy
| | - Claudia Campanella
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostic (BIND), University of Palermo, Palermo, Italy
| | - Francesca Rappa
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostic (BIND), University of Palermo, Palermo, Italy
| | - Magdalena Gorska
- Department of Medical Chemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Everly Conway de Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy.,Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD, United States
| | - Francesco Cappello
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostic (BIND), University of Palermo, Palermo, Italy.,Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | - Alberto J L Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy.,Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD, United States
| | - Antonella Marino Gammazza
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostic (BIND), University of Palermo, Palermo, Italy
| |
Collapse
|
35
|
Membrane-Associated Heat Shock Proteins in Oncology: From Basic Research to New Theranostic Targets. Cells 2020; 9:cells9051263. [PMID: 32443761 PMCID: PMC7290778 DOI: 10.3390/cells9051263] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Heat shock proteins (HSPs) constitute a large family of conserved proteins acting as molecular chaperones that play a key role in intracellular protein homeostasis, regulation of apoptosis, and protection from various stress factors (including hypoxia, thermal stress, oxidative stress). Apart from their intracellular localization, members of different HSP families such as small HSPs, HSP40, HSP60, HSP70 and HSP90 have been found to be localized on the plasma membrane of malignantly transformed cells. In the current article, the role of membrane-associated molecular chaperones in normal and tumor cells is comprehensively reviewed with implications of these proteins as plausible targets for cancer therapy and diagnostics.
Collapse
|
36
|
Liyanagamage DSNK, Martinus RD. Role of Mitochondrial Stress Protein HSP60 in Diabetes-Induced Neuroinflammation. Mediators Inflamm 2020; 2020:8073516. [PMID: 32410865 PMCID: PMC7201845 DOI: 10.1155/2020/8073516] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/10/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is the most common metabolic disorder characterized by hyperglycemia and associated malfunctions of the metabolism of carbohydrates, proteins, and lipids. There is increasing evidence of a relationship between diabetes and vascular dementia. Interestingly, hyperglycemia-linked neuroinflammation in the central nervous system is considered to play a key role during vascular dementia in diabetic patients. However, the mechanisms responsible for the relationship between hyperglycemia and neuroinflammation is not clearly understood. Diabetes-induced alternations in the blood-brain barrier permit high glucose influx into the brain cells via glucose transporters and promote oxidative stress through overproduction of reactive oxygen species. Despite many studies demonstrating a link between oxidative stress and mitochondrial dysfunction, the relationship between mitochondrial dysfunction and neuron inflammation during hyperglycemia remains to be established. In this review, we will focus on diabetes-induced changes in the central nervous system and the role of mitochondrial heat shock protein 60 (HSP60) as an initiator of oxidative stress and potential modulator of neuroinflammation. We suggest that oxidative stress-mediated mitochondrial dysfunction stimulates the upregulation of mitochondrial heat shock protein 60 (HSP60) and ultimately initiates inflammatory pathways by activating pattern recognition receptors. HSP60 also could be a focal point in the development of a biomarker of neuroinflammation as HSP60 is known to be significantly elevated in diabetic patients. Interestingly, extracellular secretion of HSP60 via exosomes suggests that inflammation could spread to neighboring astrocytes by activating pattern recognition receptors of astrocytes via neuronal exosomes containing HSP60. A mechanism for linking neuron and astrocyte inflammation will provide new therapeutic approaches to modulate neuroinflammation and therefore potentially ameliorate the cognitive impairment in diabetic brains associated with vascular dementia.
Collapse
Affiliation(s)
| | - Ryan D. Martinus
- School of Science, Division of Health, Engineering, Computing & Science, The University of Waikato, Hamilton, New Zealand
| |
Collapse
|
37
|
Role of HSP60/HSP10 in Lung Cancer: Simple Biomarkers or Leading Actors? JOURNAL OF ONCOLOGY 2020; 2020:4701868. [PMID: 32318107 PMCID: PMC7149434 DOI: 10.1155/2020/4701868] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 02/22/2020] [Accepted: 03/02/2020] [Indexed: 02/08/2023]
Abstract
Cancers are one of the major challenges faced by modern medicine both because of their impact in terms of the amount of cases and of the ineffectiveness of therapies used today. A concrete support to the fight against them can be found in the analysis and understanding of the molecular mechanisms involving molecular chaperones. In particular, HSP60 and HSP10 seem to play an important role in carcinogenesis, supporting tumours in their proliferation, survival, and metastasis. Efforts must be directed toward finding ways to eliminate or block this "mistaken" chaperone. Therefore, the scientific community must develop therapeutic strategies that consider HSP60 and HSP10 as the possible target of an anti-tumoural treatment and not only as diagnostic biomarkers, since they contribute to the evolution of pre-cancerous respiratory pathologies in lung tumours. HSP60 acts at the mitochondrial, cytoplasmic, and extracellular levels in the development of cancer pathologies. The molecular mechanisms in which these chaperones are involved concern cell survival, the restoration of a condition of absence of replicative senescence, the promotion of pro-inflammatory environments, and an increase in the ability to form metastases. In this review, we will also present examples of interactions between HSP60 and HSP10 and different molecules and ways to exploit this knowledge in anticancer therapies for lung tumours. In order to improve not only chances for an earlier diagnosis but also treatments for patients suffering from this type of disease, chaperones must be considered as key agents in carcinogenesis and primary targets in therapeutics.
Collapse
|
38
|
Curcumin Affects HSP60 Folding Activity and Levels in Neuroblastoma Cells. Int J Mol Sci 2020; 21:ijms21020661. [PMID: 31963896 PMCID: PMC7013437 DOI: 10.3390/ijms21020661] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 02/06/2023] Open
Abstract
The fundamental challenge in fighting cancer is the development of protective agents able to interfere with the classical pathways of malignant transformation, such as extracellular matrix remodeling, epithelial–mesenchymal transition and, alteration of protein homeostasis. In the tumors of the brain, proteotoxic stress represents one of the main triggering agents for cell transformation. Curcumin is a natural compound with anti-inflammatory and anti-cancer properties with promising potential for the development of therapeutic drugs for the treatment of cancer as well as neurodegenerative diseases. Among the mediators of cancer development, HSP60 is a key factor for the maintenance of protein homeostasis and cell survival. High HSP60 levels were correlated, in particular, with cancer development and progression, and for this reason, we investigated the ability of curcumin to affect HSP60 expression, localization, and post-translational modifications using a neuroblastoma cell line. We have also looked at the ability of curcumin to interfere with the HSP60/HSP10 folding machinery. The cells were treated with 6, 12.5, and 25 µM of curcumin for 24 h, and the flow cytometry analysis showed that the compound induced apoptosis in a dose-dependent manner with a higher percentage of apoptotic cells at 25 µM. This dose of curcumin-induced a decrease in HSP60 protein levels and an upregulation of HSP60 mRNA expression. Moreover, 25 µM of curcumin reduced HSP60 ubiquitination and nitration, and the chaperonin levels were higher in the culture media compared with the untreated cells. Furthermore, curcumin at the same dose was able to favor HSP60 folding activity. The reduction of HSP60 levels, together with the increase in its folding activity and the secretion in the media led to the supposition that curcumin might interfere with cancer progression with a protective mechanism involving the chaperonin.
Collapse
|
39
|
Mohammadi S, Yousefi F, Shabaninejad Z, Movahedpour A, Mahjoubin Tehran M, Shafiee A, Moradizarmehri S, Hajighadimi S, Savardashtaki A, Mirzaei H. Exosomes and cancer: From oncogenic roles to therapeutic applications. IUBMB Life 2019; 72:724-748. [PMID: 31618516 DOI: 10.1002/iub.2182] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022]
Abstract
Exosomes belong to extracellular vehicles that were produced and secreted from most eukaryotic cells and are involved in cell-to-cell communications. They are an effective delivery system for biological compounds such as mRNAs, microRNAs (miRNAs), proteins, lipids, saccharides, and other physiological compounds to target cells. In this way, they could influence on cellular pathways and mediate their physiological behaviors including cell proliferation, tumorigenesis, differentiation, and so on. Many research studies focused on their role in cancers and also on potentially therapeutic and biomarker applications. In the current study, we reviewed the exosomes' effects on cancer progression based on their cargoes including miRNAs, long noncoding RNAs, circular RNAs, DNAs, mRNAs, proteins, and lipids. Moreover, their therapeutic roles in cancer were considered. In this regard, we have given a brief overview of challenges and obstacles in using exosomes as therapeutic agents.
Collapse
Affiliation(s)
- Soheila Mohammadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Yousefi
- Department of Biological Sciences, Faculty of Genetics, Tarbiat Modares University, Tehran, Iran
| | - Zahra Shabaninejad
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.,Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Mahjoubin Tehran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alimohammad Shafiee
- Division of General Internal Medicine, Toronto General Hospital, Toronto, Canada
| | - Sanaz Moradizarmehri
- Division of General Internal Medicine, Toronto General Hospital, Toronto, Canada
| | - Sarah Hajighadimi
- Division of General Internal Medicine, Toronto General Hospital, Toronto, Canada
| | - Amir Savardashtaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
40
|
Kamm A, Przychodzeń P, Kuban–Jankowska A, Marino Gammazza A, Cappello F, Daca A, Żmijewski MA, Woźniak M, Górska–Ponikowska M. 2-Methoxyestradiol and Its Combination with a Natural Compound, Ferulic Acid, Induces Melanoma Cell Death via Downregulation of Hsp60 and Hsp90. JOURNAL OF ONCOLOGY 2019; 2019:9293416. [PMID: 32082378 PMCID: PMC7012217 DOI: 10.1155/2019/9293416] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/28/2019] [Accepted: 09/04/2019] [Indexed: 12/15/2022]
Abstract
Melanoma is an aggressive type of skin cancer with one of the highest mortality rates. Notably, its incidence in the last few decades has increased faster than any other cancer. Therefore, searching for novel anticancer therapies is of great clinical importance. In the present study, we investigated the anticancer potential of 2-methoxyestradiol, potent chemotherapeutic, in the A375 melanoma cellular model. In order to furthermore evaluate the anticancer efficacy of 2-methoxyestradiol, we have additionally combined the treatment with a naturally occurring polyphenol, ferulic acid. The results were obtained using the melanoma A375 cellular model. In the study, we used MTT assay, flow cytometry, and western blot techniques. Herein, we have evidenced that the molecular mechanism of action of 2-methoxyestradiol and ferulic acid is partly related to the reduction of Hsp60 and Hsp90 levels and the induction of nitric oxide in the A375 melanoma cell model, while no changes were observed in Hsp70 expression after 2-methoxyestradiol and ferulic acid treatment separately or in combination. This is especially important in case of chemoresistance mechanisms because the accumulation of Hsp70 reduces induction of cancer cell death, thus decreasing antitumour efficacy.
Collapse
Affiliation(s)
- Anna Kamm
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk 80-211, Poland
| | - Paulina Przychodzeń
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk 80-211, Poland
| | | | - Antonella Marino Gammazza
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Francesco Cappello
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Agnieszka Daca
- Department of Pathology and Rheumatology, Medical University of Gdansk, Gdansk 80-211, Poland
| | - Michał A. Żmijewski
- Department of Histology, Medical University of Gdansk, Gdansk 80-211, Poland
| | - Michał Woźniak
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk 80-211, Poland
| | - Magdalena Górska–Ponikowska
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk 80-211, Poland
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
41
|
Jones LB, Kumar S, Curry AJ, Price JS, Krendelchtchikov A, Crenshaw BJ, Bell CR, Williams SD, Tolliver TA, Saldanha SN, Sims B, Matthews QL. Alcohol Exposure Impacts the Composition of HeLa-Derived Extracellular Vesicles. Biomedicines 2019; 7:biomedicines7040078. [PMID: 31574936 PMCID: PMC6966524 DOI: 10.3390/biomedicines7040078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/11/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles are nanosized vesicles that are under intense investigation for their role in intercellular communication. Extracellular vesicles have begun to be examined for their role in disease protection and their role as disease biomarkers and/or vaccine agents. The goal of this study was to investigate the effects of alcohol exposure on the biogenesis and composition of extracellular vesicles derived from the cervical cancer line, HeLa. The HeLa cells were cultured in exosome-free media and were either mock-treated (control) or treated with 50 mM or 100 mM of alcohol for 24 h and 48 h. Our results demonstrated that alcohol significantly impacts HeLa cell viability and exosome biogenesis/composition. Importantly, our studies demonstrate the critical role of alcohol on HeLa cells, as well as HeLa-derived extracellular vesicle biogenesis and composition. Specifically, these results indicate that alcohol alters extracellular vesicles’ packaging of heat shock proteins and apoptotic proteins. Extracellular vesicles serve as communicators for HeLa cells, as well as biomarkers for the initiation and progression of disease.
Collapse
Affiliation(s)
- Leandra B Jones
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA.
| | - Sanjay Kumar
- Departments of Pediatrics and Cell, Developmental and Integrative Biology, Division of Neonatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Aliyah J Curry
- Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA.
- Center for Nanobiotechnology Research (CNBR), Alabama State University, Montgomery, AL 36104, USA.
| | - Jayde S Price
- Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA.
- Center for Nanobiotechnology Research (CNBR), Alabama State University, Montgomery, AL 36104, USA.
| | - Alexandre Krendelchtchikov
- Departments of Pediatrics and Cell, Developmental and Integrative Biology, Division of Neonatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Brennetta J Crenshaw
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA.
| | - Courtnee' R Bell
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA.
| | - Sparkle D Williams
- Departments of Pediatrics and Cell, Developmental and Integrative Biology, Division of Neonatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Tambre A Tolliver
- Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA.
| | - Sabita N Saldanha
- Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA.
| | - Brian Sims
- Departments of Pediatrics and Cell, Developmental and Integrative Biology, Division of Neonatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Qiana L Matthews
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA.
- Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA.
| |
Collapse
|
42
|
Caruso Bavisotto C, Cipolla C, Graceffa G, Barone R, Bucchieri F, Bulone D, Cabibi D, Campanella C, Marino Gammazza A, Pitruzzella A, Porcasi R, San Biagio PL, Tomasello G, Conway de Macario E, Macario AJL, Cappello F, Rappa F. Immunomorphological Pattern of Molecular Chaperones in Normal and Pathological Thyroid Tissues and Circulating Exosomes: Potential Use in Clinics. Int J Mol Sci 2019; 20:ijms20184496. [PMID: 31514388 PMCID: PMC6770414 DOI: 10.3390/ijms20184496] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/31/2019] [Accepted: 09/08/2019] [Indexed: 02/07/2023] Open
Abstract
The thyroid is a major component of the endocrine system and its pathology can cause serious diseases, e.g., papillary carcinoma (PC). However, the carcinogenic mechanisms are poorly understood and clinical useful biomarkers are scarce. Therefore, we determined if there are quantitative patterns of molecular chaperones in the tumor tissue and circulating exosomes that may be useful in diagnosis and provide clues on their participation in carcinogenesis. Hsp27, Hsp60, Hsp70, and Hsp90 were quantified by immunohistochemistry in PC, benign goiter (BG), and normal peritumoral tissue (PT). The same chaperones were assessed in plasma exosomes from PC and BG patients before and after ablative surgery, using Western blotting. Hsp27, Hsp60, and Hsp90 were increased in PC in comparison with PT and BG but no differences were found for Hsp70. Similarly, exosomal levels of Hsp27, Hsp60, and Hsp90 were higher in PC than in BG, and those in PC were higher before ablative surgery than after it. Hsp27, Hsp60, and Hsp90 show distinctive quantitative patterns in thyroid tissue and circulating exosomes in PC as compared with BG, suggesting some implication in the carcinogenesis of these chaperones and indicating their potential as biomarkers for clinical applications.
Collapse
Affiliation(s)
- Celeste Caruso Bavisotto
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90100 Palermo, Italy.
- Institute of Biophysics, National Research Council, 90100 Palermo, Italy.
| | - Calogero Cipolla
- Department of Surgical Oncology and Oral Sciences, University of Palermo, 90127 Palermo, Italy.
| | - Giuseppa Graceffa
- Department of Surgical Oncology and Oral Sciences, University of Palermo, 90127 Palermo, Italy.
| | - Rosario Barone
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy.
| | - Fabio Bucchieri
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy.
- Institute of Biomedicine and Molecular Immunology, National Research Council, 90100 Palermo, Italy.
| | - Donatella Bulone
- Institute of Biophysics, National Research Council, 90100 Palermo, Italy.
| | - Daniela Cabibi
- Department "G. D'Alessandro", Pathology Institute, University of Palermo, 90127 Palermo, Italy.
| | - Claudia Campanella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy.
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy.
| | - Alessandro Pitruzzella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy.
| | - Rossana Porcasi
- Department "G. D'Alessandro", Pathology Institute, University of Palermo, 90127 Palermo, Italy.
| | | | - Giovanni Tomasello
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy.
| | - Everly Conway de Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90100 Palermo, Italy.
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA.
| | - Alberto J L Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90100 Palermo, Italy.
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA.
| | - Francesco Cappello
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90100 Palermo, Italy.
| | - Francesca Rappa
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy.
| |
Collapse
|
43
|
Vilasi S, Carrotta R, Ricci C, Rappa GC, Librizzi F, Martorana V, Ortore MG, Mangione MR. Inhibition of Aβ 1-42 Fibrillation by Chaperonins: Human Hsp60 Is a Stronger Inhibitor than Its Bacterial Homologue GroEL. ACS Chem Neurosci 2019; 10:3565-3574. [PMID: 31298838 DOI: 10.1021/acschemneuro.9b00183] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease is a chronic neurodegenerative disease characterized by the accumulation of pathological aggregates of amyloid beta peptide. Many efforts have been focused on understanding peptide aggregation pathways and on identification of molecules able to inhibit aggregation in order to find an effective therapy. As a result, interest in neuroprotective proteins, such as molecular chaperones, has increased as their normal function is to assist in protein folding or to facilitate the disaggregation and/or clearance of abnormal aggregate proteins. Using biophysical techniques, we evaluated the effects of two chaperones, human Hsp60 and bacterial GroEL, on the fibrillogenesis of Aβ1-42. Both chaperonins interfere with Aβ1-42 aggregation, but the effect of Hsp60 is more significant and correlates with its more pronounced flexibility and stronger interaction with ANS, an indicator of hydrophobic regions. Dose-dependent ThT fluorescence kinetics and SAXS experiments reveal that Hsp60 does not change the nature of the molecular processes stochastically leading to the formation of seeds, but strongly delays them by recognition of hydrophobic sites of some peptide species crucial for triggering amyloid formation. Hsp60 reduces the initial chaotic heterogeneity of Aβ1-42 sample at high concentration regimes. The understanding of chaperone action in counteracting pathological aggregation could be a starting point for potential new therapeutic strategies against neurodegenerative diseases.
Collapse
Affiliation(s)
- Silvia Vilasi
- Institute of Biophysics, National Research Council, Palermo 90146, Italy
| | - Rita Carrotta
- Institute of Biophysics, National Research Council, Palermo 90146, Italy
| | - Caterina Ricci
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona 60131, Italy
| | | | - Fabio Librizzi
- Institute of Biophysics, National Research Council, Palermo 90146, Italy
| | - Vincenzo Martorana
- Institute of Biophysics, National Research Council, Palermo 90146, Italy
| | - Maria Grazia Ortore
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona 60131, Italy
| | | |
Collapse
|
44
|
Lin J, Xia L, Liang J, Han Y, Wang H, Oyang L, Tan S, Tian Y, Rao S, Chen X, Tang Y, Su M, Luo X, Wang Y, Wang H, Zhou Y, Liao Q. The roles of glucose metabolic reprogramming in chemo- and radio-resistance. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:218. [PMID: 31122265 PMCID: PMC6533757 DOI: 10.1186/s13046-019-1214-z] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/07/2019] [Indexed: 12/15/2022]
Abstract
Reprogramming of cancer metabolism is a newly recognized hallmark of malignancy. The aberrant glucose metabolism is associated with dramatically increased bioenergetics, biosynthetic, and redox demands, which is vital to maintain rapid cell proliferation, tumor progression, and resistance to chemotherapy and radiation. When the glucose metabolism of cancer is rewiring, the characters of cancer will also occur corresponding changes to regulate the chemo- and radio-resistance of cancer. The procedure is involved in the alteration of many activities, such as the aberrant DNA repairing, enhanced autophagy, oxygen-deficient environment, and increasing exosomes secretions, etc. Targeting altered metabolic pathways related with the glucose metabolism has become a promising anti-cancer strategy. This review summarizes recent progress in our understanding of glucose metabolism in chemo- and radio-resistance malignancy, and highlights potential molecular targets and their inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Jinguan Lin
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Longzheng Xia
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jiaxin Liang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yaqian Han
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Heran Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Linda Oyang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Shiming Tan
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yutong Tian
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Shan Rao
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xiaoyan Chen
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yanyan Tang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Min Su
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xia Luo
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Ying Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Hui Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yujuan Zhou
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Qianjin Liao
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
45
|
Sabharwal P, Amritha CK, Sushmitha C, Natraj U, Savithri HS. Intracellular trafficking and endocytic uptake pathway of Pepper vein banding virus-like particles in epithelial cells. Nanomedicine (Lond) 2019; 14:1247-1265. [PMID: 31084385 DOI: 10.2217/nnm-2018-0405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aim: Plant virus-like particles (VLPs) have emerged as a novel platform for delivery of drugs/antibodies. The aim of the present investigation is to establish the entry mechanism of flexuous rod-shaped virus particles into mammalian cells. Methods: Far-Western blot analysis, pull-down and ELISA were used to characterize vimentin and Hsp60 interaction with VLPs. The mode/kinetics of internalization of VLPs was deciphered using pharmacological inhibitors/endosomal markers. Results & discussion: The flexuous rod-shaped VLPs of Pepper vein banding virus (PVBV) enter HeLa and HepG2 cells via cell-surface proteins: vimentin and Hsp60, respectively. VLPs internalize via different modes of endocytosis in HeLa, HepG2 cells and are biodegradable. Vimentin and Hsp60 could be potential epithelial ligands that facilitate targeting of nanoparticles to tumor cells.
Collapse
Affiliation(s)
- Pallavi Sabharwal
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | | | - Cheekati Sushmitha
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Usha Natraj
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | | |
Collapse
|
46
|
Extracellular Vesicle-Mediated Cell⁻Cell Communication in the Nervous System: Focus on Neurological Diseases. Int J Mol Sci 2019; 20:ijms20020434. [PMID: 30669512 PMCID: PMC6359416 DOI: 10.3390/ijms20020434] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/12/2019] [Accepted: 01/17/2019] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), including exosomes, are membranous particles released by cells into the extracellular space. They are involved in cell differentiation, tissue homeostasis, and organ remodelling in virtually all tissues, including the central nervous system (CNS). They are secreted by a range of cell types and via blood reaching other cells whose functioning they can modify because they transport and deliver active molecules, such as proteins of various types and functions, lipids, DNA, and miRNAs. Since they are relatively easy to isolate, exosomes can be characterized, and their composition elucidated and manipulated by bioengineering techniques. Consequently, exosomes appear as promising theranostics elements, applicable to accurately diagnosing pathological conditions, and assessing prognosis and response to treatment in a variety of disorders. Likewise, the characteristics and manageability of exosomes make them potential candidates for delivering selected molecules, e.g., therapeutic drugs, to specific target tissues. All these possible applications are pertinent to research in neurophysiology, as well as to the study of neurological disorders, including CNS tumors, and autoimmune and neurodegenerative diseases. In this brief review, we discuss what is known about the role and potential future applications of exosomes in the nervous system and its diseases, focusing on cell–cell communication in physiology and pathology.
Collapse
|
47
|
Fais S, Logozzi M, Alberti G, Campanella C. Exosomal Hsp60: A Tumor Biomarker? HEAT SHOCK PROTEIN 60 IN HUMAN DISEASES AND DISORDERS 2019. [DOI: 10.1007/978-3-030-23154-5_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
48
|
Cappello F, Mazzola M, Jurjus A, Zeenny MN, Jurjus R, Carini F, Leone A, Bonaventura G, Tomasello G, Bucchieri F, Conway de Macario E, Macario AJL. Hsp60 as a Novel Target in IBD Management: A Prospect. Front Pharmacol 2019; 10:26. [PMID: 30800066 PMCID: PMC6376446 DOI: 10.3389/fphar.2019.00026] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 01/10/2019] [Indexed: 12/16/2022] Open
Abstract
Inflammatory bowel disease (IBD) encompasses various pathological conditions similar but distinct that share a multifactorial etiology, including involvement of the intestinal barrier function, the immune system, and intestinal microorganisms. Hsp60 is a chaperonin component of the chaperoning system, present in all cells and tissues, including the intestine. It plays important roles in cell physiology outside and inside mitochondria, its canonical place of residence. However, Hsp60 can also be pathogenic in many conditions, the Hsp60 chaperonopathies, possibly including IBD. The various clinico-pathological types of IBD have a complicated mix of causative factors, among which Hsp60 can be considered a putatively important driver of events and could play an etiopathogenic role. This possibility is discussed in this review. We also indicate that Hsp60 can be a biomarker useful in disease diagnosing and monitoring and, if found active in pathogenesis, should become a target for developing new therapies. The latter are particularly needed to alleviate patient suffering and to prevent complications, including colon cancer.
Collapse
Affiliation(s)
- Francesco Cappello
- Department of Experimental Biomedicine and Clinical Neuroscience University of Palermo (BIONEC-UniPA), Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States
- *Correspondence: Francesco Cappello,
| | - Margherita Mazzola
- Department of Experimental Biomedicine and Clinical Neuroscience University of Palermo (BIONEC-UniPA), Palermo, Italy
| | - Abdo Jurjus
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut, Beirut, Lebanon
| | - Marie-Noel Zeenny
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut, Beirut, Lebanon
| | - Rosalyn Jurjus
- Department of Anatomy and Cell Biology, Faculty Development Associate for Education Research, Center for Faculty Excellence, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Francesco Carini
- Department of Experimental Biomedicine and Clinical Neuroscience University of Palermo (BIONEC-UniPA), Palermo, Italy
| | - Angelo Leone
- Department of Experimental Biomedicine and Clinical Neuroscience University of Palermo (BIONEC-UniPA), Palermo, Italy
| | - Giuseppe Bonaventura
- Department of Experimental Biomedicine and Clinical Neuroscience University of Palermo (BIONEC-UniPA), Palermo, Italy
| | - Giovanni Tomasello
- Department of Experimental Biomedicine and Clinical Neuroscience University of Palermo (BIONEC-UniPA), Palermo, Italy
| | - Fabio Bucchieri
- Department of Experimental Biomedicine and Clinical Neuroscience University of Palermo (BIONEC-UniPA), Palermo, Italy
| | - Everly Conway de Macario
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore – Institute of Marine and Environmental Technology (IMET), Baltimore, MD, United States
| | - Alberto J. L. Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore – Institute of Marine and Environmental Technology (IMET), Baltimore, MD, United States
| |
Collapse
|
49
|
Caruso Bavisotto C, Graziano F, Rappa F, Marino Gammazza A, Logozzi M, Fais S, Maugeri R, Bucchieri F, Conway de Macario E, Macario AJL, Cappello F, Iacopino DG, Campanella C. Exosomal Chaperones and miRNAs in Gliomagenesis: State-of-Art and Theranostics Perspectives. Int J Mol Sci 2018; 19:E2626. [PMID: 30189598 PMCID: PMC6164348 DOI: 10.3390/ijms19092626] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/19/2022] Open
Abstract
Gliomas have poor prognosis no matter the treatment applied, remaining an unmet clinical need. As background for a substantial change in this situation, this review will focus on the following points: (i) the steady progress in establishing the role of molecular chaperones in carcinogenesis; (ii) the recent advances in the knowledge of miRNAs in regulating gene expression, including genes involved in carcinogenesis and genes encoding chaperones; and (iii) the findings about exosomes and their cargo released by tumor cells. We would like to trigger a discussion about the involvement of exosomal chaperones and miRNAs in gliomagenesis. Chaperones may be either targets for therapy, due to their tumor-promoting activity, or therapeutic agents, due to their antitumor growth activity. Thus, chaperones may well represent a Janus-faced approach against tumors. This review focuses on extracellular chaperones as part of exosomes' cargo, because of their potential as a new tool for the diagnosis and management of gliomas. Moreover, since exosomes transport chaperones and miRNAs (the latter possibly related to chaperone gene expression in the recipient cell), and probably deliver their cargo in the recipient cells, a new area of investigation is now open, which is bound to generate significant advances in the understanding and treatment of gliomas.
Collapse
Affiliation(s)
- Celeste Caruso Bavisotto
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
- Institute of Biophysics, National Research Council, 90143 Palermo, Italy.
| | - Francesca Graziano
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Neurosurgery, University of Palermo, 90127 Palermo, Italy.
| | - Francesca Rappa
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
| | - Antonella Marino Gammazza
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
| | - Mariantonia Logozzi
- Department of Oncology and Molecular Medicine, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Rosario Maugeri
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Neurosurgery, University of Palermo, 90127 Palermo, Italy.
| | - Fabio Bucchieri
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
| | - Everly Conway de Macario
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA.
| | - Alberto J L Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA.
| | - Francesco Cappello
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
| | - Domenico G Iacopino
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Neurosurgery, University of Palermo, 90127 Palermo, Italy.
| | - Claudia Campanella
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
| |
Collapse
|
50
|
Lee H, Zhang D, Laskin DL, Jin Y. Functional Evidence of Pulmonary Extracellular Vesicles in Infectious and Noninfectious Lung Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:1500-1509. [PMID: 29997122 PMCID: PMC6109965 DOI: 10.4049/jimmunol.1800264] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 06/25/2018] [Indexed: 01/08/2023]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a highly complex process that can be triggered by both noninfectious (sterile) and infectious stimuli. Inflammatory lung responses are one of the key features in the pathogenesis of this devastating syndrome. How ALI/ARDS-associated inflammation develops remains incompletely understood, particularly after exposure to sterile stimuli. Emerging evidence suggests that extracellular vesicles (EVs) regulate intercellular communication and inflammatory responses in various diseases. In this study, we characterized the generation and function of pulmonary EVs in the setting of ALI/ARDS, induced by sterile stimuli (oxidative stress or acid aspiration) and infection (LPS/Gram-negative bacteria) in mice. EVs detected in bronchoalveolar lavage fluid (BALF) were markedly increased after exposure of animals to both types of stimuli. After sterile stimuli, alveolar type-І epithelial cells were the main source of the BALF EVs. In contrast, infectious stimuli-induced BALF EVs were mainly derived from alveolar macrophages (AMs). Functionally, BALF EVs generated in both the noninfectious and infectious ALI models promoted the recruitment of macrophages in in vivo mouse models. Furthermore, BALF EVs differentially regulated AM production of cytokines and inflammatory mediators, as well as TLR expression in AMs in vivo. Regardless of their origin, BALF EVs contributed significantly to the development of lung inflammation in both the sterile and infectious ALI. Collectively, our results provide novel insights into the mechanisms by which EVs regulate the development of lung inflammation in response to diverse stimuli, potentially providing novel therapeutic and diagnostic targets for ALI/ARDS.
Collapse
Affiliation(s)
- Heedoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118; and
| | - Duo Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118; and
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118; and
| |
Collapse
|