1
|
de Azevedo ALK, Gomig THB, Ribeiro EMDSF. Stress-induced phosphoprotein 1: how does this co-chaperone influence the metastasis steps? Clin Exp Metastasis 2024; 41:589-597. [PMID: 38581620 DOI: 10.1007/s10585-024-10282-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/27/2024] [Indexed: 04/08/2024]
Abstract
In several cancer types, metastasis is associated with poor prognosis, survival, and quality of life, representing a life risk more significant than the primary tumor itself. Metastasis is a multi-step process that spreads tumor cells from primary sites to surrounding or distant organs, originating secondary tumors. The interconnected steps that drive metastasis depend of several capabilities that enable cells to detach from the primary tumor, acquire motility and migrate through the basal membrane; invade and spread through the vascular system, and finally settle and originate a new tumor. Recently, stress-induced phosphoprotein 1 (STIP1) has emerged as a protein capable of driving tumor cells through these metastasis steps by mediating several biological processes and signaling pathways. This protein is mainly known for its function as a co-chaperone, acting as a scaffold for the interaction of its client heat-shock proteins Hsp70/90 chaperones; however, it is also known that STIP1 can act independently of chaperones to activate downstream phosphorylation pathways. The over-expression of STIP1 has been reported across various cancer types, identifying it as a potential biomarker for predicting patient prognosis and monitoring the progression of metastasis. Here, we present a discussion on how this co-chaperone mediates the initial steps of metastasis (cell adhesion loss, epithelial-to-mesenchymal transition, and angiogenesis), highlighting the biological mechanisms in which STIP1 plays a vital role, also presenting an overview of the current knowledge regarding its clinical relevance.
Collapse
Affiliation(s)
- Alexandre Luiz Korte de Azevedo
- Genetics Post-Graduation Program, Genetics Department, Federal University of Paraná, P.O. box 19071, Curitiba, Paraná, CEP: 81531-990, Brazil
| | - Talita Helen Bombardelli Gomig
- Genetics Post-Graduation Program, Genetics Department, Federal University of Paraná, P.O. box 19071, Curitiba, Paraná, CEP: 81531-990, Brazil
| | | |
Collapse
|
2
|
Zhang W, Lei W, Shen F, Wang M, Li L, Chang J. Cinnamaldehyde induces apoptosis and enhances anti-colorectal cancer activity via covalent binding to HSPD1. Phytother Res 2024; 38:4957-4966. [PMID: 37086182 DOI: 10.1002/ptr.7840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/03/2023] [Accepted: 04/06/2023] [Indexed: 04/23/2023]
Abstract
Colorectal cancer (CRC) is a common malignant tumor with high morbidity and mortality rates worldwide. Although surgical resection and adjuvant radiotherapy/chemotherapy are the mainstays of CRC treatment, the efficacy is unsatisfactory due to several limitations, including high drug resistance. Accordingly, there is a dire need for new drugs or a novel combination approach to treat this patient population. Herein, we found that cinnamaldehyde (CA) could exert an antitumor effect in HCT-116 cell lines. Target fishing, molecular imaging, and live-cell tracing using an alkynyl-CA probe revealed that the heat shock 60 kDa protein 1 (HSPD1) protein was the target of CA. The covalent binding of CA with HSPD1 altered its stability. Furthermore, our results demonstrated that CA could induce cell apoptosis by inhibiting the PI3K/Akt signaling pathway and enhanced anti-CRC activity both in vitro and in vivo. Meanwhile, CA combined with different chemotherapeutic agents was beneficial to patients resistant to anti-CRC drug therapy.
Collapse
Affiliation(s)
- Weiyi Zhang
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology (XJDX1713), School of Pharmacy, Xinjiang Medical University, Urumchi, China
| | - Wei Lei
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fukui Shen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Mukuo Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Linlin Li
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology (XJDX1713), School of Pharmacy, Xinjiang Medical University, Urumchi, China
| | - Junmin Chang
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology (XJDX1713), School of Pharmacy, Xinjiang Medical University, Urumchi, China
| |
Collapse
|
3
|
Nabih HK, Yücer R, Mahmoud N, Dawood M, Elbadawi M, Shahhamzehei N, Atia MAM, AbdelSadik A, Hussien TA, Ibrahim MAA, Klauck SM, Hegazy MEF, Efferth T. The cytotoxic activities of the major diterpene extracted from Salvia multicaulis (Bardakosh) are mediated by the regulation of heat-shock response and fatty acid metabolism pathways in human leukemia cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156023. [PMID: 39368339 DOI: 10.1016/j.phymed.2024.156023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 07/15/2024] [Accepted: 09/02/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Leukemia is one of the most lethal cancers worldwide and represents the sixth-leading cause of cancer deaths. The results of leukemia treatment have not been as positive as desired, and recurrence is common. PURPOSE Thus, there is an urgent requirement for the development of new therapeutic drugs. Salvia multicaulis (Bardakosh) is a widespread species that contains multiple phytochemical components with anti-cancer activities. METHODS We isolated and characterized the major diterpene candesalvone B methyl ester from S. multicaulis and investigated its action as a cytotoxic agent towards sensitive and drug-resistant leukemia cells by the resazurin reduction assay. Additionally, the targeted genes and the affected molecular mechanisms attributed to the potent cytotoxic activities were discovered by transcriptome-wide mRNA expression profiling. The targets predicted to be regulated by candesalvone B methyl ester in each cell line were confirmed by qRT-PCR, molecular docking, microscale thermophoresis, and western blotting. Moreover, cell cycle distribution and apoptosis were analyzed by flow cytometry. RESULTS Candesalvone B methyl ester was cytotoxic with IC50 values of 20.95 ± 0.15 µM against CCRF-CEM cells and 4.13 ± 0.10 µM against multidrug-resistant CEM/ADR5000 leukemia cells. The pathway enrichment analysis disclosed that candesalvone B methyl ester could regulate the heat-shock response signaling pathway via targeting heat shock factor 1 (HSF1) in CCRF-CEM cells and ELOVL fatty acid elongase 5 (ELOVL5) controls the fatty acid metabolism pathway in CEM/ADR5000 cells. Microscale thermophoresis showed the binding of candesalvone B methyl ester with HSF1 and ELOVL5, confirming the results of molecular docking analysis. Down-regulation of both HSF1 and ELOVL5 by candesalvone B methyl ester as detected by both western blotting and RT-qPCR was related to the reversal of drug resistance in the leukemia cells. Furthermore, candesalvone B methyl ester increased the arrest in the sub-G1 phase of the cell cycle in a dose-dependent manner from 1.3 % to 32.3 % with concomitant induction of apoptosis up to 29.0 % in CCRF-CEM leukemic cells upon inhibition of HSF1. CONCLUSION Candesalvone B methyl ester isolated from S. multicaulis exerted cytotoxicity by affecting apoptosis, cell division, and modulation of expression levels of genes contributing to the heat stress signaling and fatty acid metabolism pathways that could relieve drug resistance of leukemia cells.
Collapse
Affiliation(s)
- Heba K Nabih
- National Research Centre, Medical Biochemistry Department, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Rümeysa Yücer
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Nuha Mahmoud
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Mona Dawood
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; Department of Molecular Biology, Faculty of Medical Laboratory Science, Al-Neelain University, Khartoum, Sudan
| | - Mohamed Elbadawi
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Nasim Shahhamzehei
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Mohamed A M Atia
- Molecular Genetics and Genome Mapping Laboratory, Genome Mapping Department, Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center (ARC), Giza 12619, Egypt
| | - Ahmed AbdelSadik
- Zoology Department, Faculty of Science, Aswan University, 81528 Aswan, Egypt; Molecular Biotechnology Program, Faculty of Advanced Basic Sciences, Galala University, 43552, New Galala, Egypt
| | - Taha A Hussien
- Pharmacognosy Department, Faculty of Pharmacy, Sphinx University, New Assiut City, Assiut 10, Egypt
| | - Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia 61519, Egypt; School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Sabine M Klauck
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Mohamed-Elamir F Hegazy
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; National Research Centre, Chemistry of Medicinal Plants Department, 33 El-Bohouth St., Dokki, Giza 12622, Egypt.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
4
|
Zuo WF, Pang Q, Zhu X, Yang QQ, Zhao Q, He G, Han B, Huang W. Heat shock proteins as hallmarks of cancer: insights from molecular mechanisms to therapeutic strategies. J Hematol Oncol 2024; 17:81. [PMID: 39232809 PMCID: PMC11375894 DOI: 10.1186/s13045-024-01601-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
Heat shock proteins are essential molecular chaperones that play crucial roles in stabilizing protein structures, facilitating the repair or degradation of damaged proteins, and maintaining proteostasis and cellular functions. Extensive research has demonstrated that heat shock proteins are highly expressed in cancers and closely associated with tumorigenesis and progression. The "Hallmarks of Cancer" are the core features of cancer biology that collectively define a series of functional characteristics acquired by cells as they transition from a normal state to a state of tumor growth, including sustained proliferative signaling, evasion of growth suppressors, resistance to cell death, enabled replicative immortality, the induction of angiogenesis, and the activation of invasion and metastasis. The pivotal roles of heat shock proteins in modulating the hallmarks of cancer through the activation or inhibition of various signaling pathways has been well documented. Therefore, this review provides an overview of the roles of heat shock proteins in vital biological processes from the perspective of the hallmarks of cancer and summarizes the small-molecule inhibitors that target heat shock proteins to regulate various cancer hallmarks. Moreover, we further discuss combination therapy strategies involving heat shock proteins and promising dual-target inhibitors to highlight the potential of targeting heat shock proteins for cancer treatment. In summary, this review highlights how targeting heat shock proteins could regulate the hallmarks of cancer, which will provide valuable information to better elucidate and understand the roles of heat shock proteins in oncology and the mechanisms of cancer occurrence and development and aid in the development of more efficacious and less toxic novel anticancer agents.
Collapse
Affiliation(s)
- Wei-Fang Zuo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qiwen Pang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xinyu Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qian-Qian Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qian Zhao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Gu He
- Department of Dermatology and Venereology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
5
|
Shi R, Yu R, Lian F, Zheng Y, Feng S, Li C, Zheng X. Targeting HSP47 for cancer treatment. Anticancer Drugs 2024; 35:623-637. [PMID: 38718070 DOI: 10.1097/cad.0000000000001612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Heat shock protein 47 (HSP47) serves as an endoplasmic reticulum residing collagen-specific chaperone and plays an important role in collagen biosynthesis and structural assembly. HSP47 is encoded by the SERPINH1 gene, which is located on chromosome 11q13.5, one of the most frequently amplified regions in human cancers. The expression of HSP47 is regulated by multiple cellular factors, including cytokines, transcription factors, microRNAs, and circular RNAs. HSP47 is frequently upregulated in a variety of cancers and plays an important role in tumor progression. HSP47 promotes tumor stemness, angiogenesis, growth, epithelial-mesenchymal transition, and metastatic capacity. HSP47 also regulates the efficacy of tumor therapies, such as chemotherapy, radiotherapy, and immunotherapy. Inhibition of HSP47 expression has antitumor effects, suggesting that targeting HSP47 is a feasible strategy for cancer treatment. In this review, we highlight the function and expression of regulatory mechanisms of HSP47 in cancer progression and point out the potential development of therapeutic strategies in targeting HSP47 in the future.
Collapse
Affiliation(s)
- Run Shi
- School of Medicine, Pingdingshan University, Pingdingshan, China
| | | | | | | | | | | | | |
Collapse
|
6
|
Liu S, Liu Y, Bao E, Tang S. The Protective Role of Heat Shock Proteins against Stresses in Animal Breeding. Int J Mol Sci 2024; 25:8208. [PMID: 39125776 PMCID: PMC11311290 DOI: 10.3390/ijms25158208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Heat shock proteins (HSPs) play an important role in all living organisms under stress conditions by acting as molecular chaperones. The expression of different HSPs during stress varies depending on their protective functions and anti-apoptotic activities. The application of HSPs improves the efficiency and decreases the economic cost of animal breeding. By upregulating the expression of HSPs, feed supplements can improve stress tolerance in farm animals. In addition, high expression of HSPs is often a feature of tumor cells, and inhibiting the expression of HSPs is a promising novel method for killing these cells and treating cancers. In the present review, the findings of previous research on the application of HSPs in animal breeding and veterinary medicine are summarized, and the knowledge of the actions of HSPs in animals is briefly discussed.
Collapse
Affiliation(s)
| | | | - Endong Bao
- College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1 Road, Nanjing 210095, China; (S.L.); (Y.L.)
| | - Shu Tang
- College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1 Road, Nanjing 210095, China; (S.L.); (Y.L.)
| |
Collapse
|
7
|
Fandiño-Devia E, Brankiewicz A, Santa-González GA, Guevara-Lora I, Manrique-Moreno M. Comparative Study of the Potential Cell-Penetrating Peptide ∆M4 on Apoptosis Cell Signaling in A375 and A431 Cancer Cell Lines. Pharmaceutics 2024; 16:775. [PMID: 38931896 PMCID: PMC11207241 DOI: 10.3390/pharmaceutics16060775] [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: 05/05/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
In recent yearsjajajj, peptide-based therapeutics have attracted increasing interest as a potential approach to cancer treatment. Peptides are characterized by high specificity and low cytotoxicity, but they cannot be considered universal drugs for all types of cancer. Of the numerous anticancer-reported peptides, both natural and synthetic, only a few have reached clinical applications. However, in most cases, the mechanism behind the anticancer activity of the peptide is not fully understood. For this reason, in this work, we investigated the effect of the novel peptide ∆M4, which has documented anticancer activity, on two human skin cancer cell lines. A novel approach to studying the potential induction of apoptosis by anticancer peptides is the use of protein microarrays. The results of the apoptosis protein study demonstrated that both cell types, skin malignant melanoma (A375) and epidermoid carcinoma (A431), exhibited markers associated with apoptosis and cellular response to oxidative stress. Additionally, ∆M4 induced concentration- and time-dependent moderate ROS production, triggering a defensive response from the cells, which showed decreased activation of cytoplasmic superoxide dismutase. However, the studied cells exhibited a differential response in catalase activity, with A375 cells showing greater resistance to the peptide action, possibly mediated by the Nrf2 pathway. Nevertheless, both cell types showed moderate activity of caspases 3/7, suggesting that they may undergo partial apoptosis, although another pathway of programmed death cannot be excluded. Extended analysis of the mechanisms of action of anticancer peptides may help determine their effectiveness in overcoming chemoresistance in cancerous cells.
Collapse
Affiliation(s)
- Estefanía Fandiño-Devia
- Group of Structural Biochemistry of Macromolecules, Faculty of Exact and Natural Sciences, University of Antioquia, A.A. 1226, Medellin 050010, Colombia;
| | - Aleksandra Brankiewicz
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland;
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-387 Krakow, Poland
| | - Gloria A. Santa-González
- Grupo de Investigación e Innovación Biomédica, Facultad de Ciencias Exactas y Aplicadas, Instituto Tecnológico Metropolitano, A.A. 54959, Medellin 050010, Colombia;
| | - Ibeth Guevara-Lora
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland;
| | - Marcela Manrique-Moreno
- Group of Structural Biochemistry of Macromolecules, Faculty of Exact and Natural Sciences, University of Antioquia, A.A. 1226, Medellin 050010, Colombia;
| |
Collapse
|
8
|
Liu D, Zuo R, Liu W, He Y, Wang Y, Yue P, Gong W, Cui J, Zhu F, Luo Y, Qi L, Guo Y, Chen L, Li G, Liu Z, Chen P, Guo H. DNAJC24 acts directly with PCNA and promotes malignant progression of LUAD by activating phosphorylation of AKT. FASEB J 2024; 38:e23630. [PMID: 38713100 DOI: 10.1096/fj.202300667rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 03/09/2024] [Accepted: 04/12/2024] [Indexed: 05/08/2024]
Abstract
Heat shock proteins (HSPs) are a group of highly conserved proteins found in a wide range of organisms. In recent years, members of the HSP family were overexpressed in various tumors and widely involved in oncogenesis, tumor development, and therapeutic resistance. In our previous study, DNAJC24, a member of the DNAJ/HSP40 family of HSPs, was found to be closely associated with the malignant phenotype of hepatocellular carcinoma. However, its relationship with other malignancies needs to be further explored. Herein, we demonstrated that DNAJC24 exhibited upregulated expression in LUAD tissue samples and predicted poor survival in LUAD patients. The upregulation of DNAJC24 expression promoted proliferation and invasion of LUAD cells in A549 and NCI-H1299 cell lines. Further studies revealed that DNAJC24 could regulate the PI3K/AKT signaling pathway by affecting AKT phosphorylation. In addition, a series of experiments such as Co-IP and mass spectrometry confirmed that DNAJC24 could directly interact with PCNA and promoted the malignant phenotypic transformation of LUAD. In conclusion, our results suggested that DNAJC24 played an important role in the progression of LUAD and may serve as a specific prognostic biomarker for LUAD patients. The DNAJC24/PCNA/AKT axis may be a potential target for future individualized and precise treatment of LUAD patients.
Collapse
Affiliation(s)
- Dongming Liu
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ran Zuo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wei Liu
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuchao He
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yu Wang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ping Yue
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenchen Gong
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jinfang Cui
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fuyi Zhu
- Department of Oncology Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yi Luo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lisha Qi
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yan Guo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Cancer Biobank of Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Liwei Chen
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Guangtao Li
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhiyong Liu
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Peng Chen
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hua Guo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
9
|
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
|
10
|
de Boer RJ, van Lidth de Jeude JF, Heijmans J. ER stress and the unfolded protein response in gastrointestinal stem cells and carcinogenesis. Cancer Lett 2024; 587:216678. [PMID: 38360143 DOI: 10.1016/j.canlet.2024.216678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/17/2024]
Abstract
Endoplasmic reticulum (ER) stress and the adaptive response that follows, termed the unfolded protein response (UPR), are crucial molecular mechanisms to maintain cellular integrity by safeguarding proper protein synthesis. Next to being important in protein homeostasis, the UPR is intricate in cell fate decisions such as proliferation, differentiation, and stemness. In the intestine, stem cells are critical in governing epithelial homeostasis and they are the cell of origin of gastrointestinal malignancies. In this review, we will discuss the role of ER stress and the UPR in the gastrointestinal tract, focusing on stem cells and carcinogenesis. Insights in mechanisms that connect ER stress and UPR with stemness and carcinogenesis may broaden our understanding in the development of cancer throughout the gastrointestinal tract and how we can exploit these mechanisms to target these malignancies.
Collapse
Affiliation(s)
- Ruben J de Boer
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 69-71, Amsterdam, The Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Jooske F van Lidth de Jeude
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 69-71, Amsterdam, The Netherlands
| | - Jarom Heijmans
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 69-71, Amsterdam, The Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands; Amsterdam UMC, University of Amsterdam, Department of General Internal Medicine and Department of Hematology, Meibergdreef 9, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Paul R, Shreya S, Pandey S, Shriya S, Abou Hammoud A, Grosset CF, Prakash Jain B. Functions and Therapeutic Use of Heat Shock Proteins in Hepatocellular Carcinoma. LIVERS 2024; 4:142-163. [DOI: 10.3390/livers4010011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
Heat shock proteins are intracellular proteins expressed in prokaryotes and eukaryotes that help protect the cell from stress. They play an important role in regulating cell cycle and cell death, work as molecular chaperons during the folding of newly synthesized proteins, and also in the degradation of misfolded proteins. They are not only produced under stress conditions like acidosis, energy depletion, and oxidative stress but are also continuously synthesized as a result of their housekeeping functions. There are different heat shock protein families based on their molecular weight, like HSP70, HSP90, HSP60, HSP27, HSP40, etc. Heat shock proteins are involved in many cancers, particularly hepatocellular carcinoma, the main primary tumor of the liver in adults. Their deregulations in hepatocellular carcinoma are associated with metastasis, angiogenesis, cell invasion, and cell proliferation and upregulated heat shock proteins can be used as either diagnostic or prognostic markers. Targeting heat shock proteins is a relevant strategy for the treatment of patients with liver cancer. In this review, we provide insights into heat shock proteins and heat shock protein-like proteins (clusterin) in the progression of hepatocellular carcinoma and their use as therapeutic targets.
Collapse
Affiliation(s)
- Ramakrushna Paul
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Smriti Shreya
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| | | | - Srishti Shriya
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Aya Abou Hammoud
- MIRCADE Team, U1312, Bordeaux Institute of Oncology, BRIC, INSERM, University of Bordeaux, 33000 Bordeaux, France
| | - Christophe F. Grosset
- MIRCADE Team, U1312, Bordeaux Institute of Oncology, BRIC, INSERM, University of Bordeaux, 33000 Bordeaux, France
| | - Buddhi Prakash Jain
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| |
Collapse
|
12
|
Viana P, Hamar P. Targeting the heat shock response induced by modulated electro-hyperthermia (mEHT) in cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189069. [PMID: 38176599 DOI: 10.1016/j.bbcan.2023.189069] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
The Heat Shock Response (HSR) is a cellular stress reaction crucial for cell survival against stressors, including heat, in both healthy and cancer cells. Modulated electro-hyperthermia (mEHT) is an emerging non-invasive cancer therapy utilizing electromagnetic fields to selectively target cancer cells via temperature-dependent and independent mechanisms. However, mEHT triggers HSR in treated cells. Despite demonstrated efficacy in cancer treatment, understanding the underlying molecular mechanisms for improved therapeutic outcomes remains a focus. This review examines the HSR induced by mEHT in cancer cells, discussing potential strategies to modulate it for enhanced tumor-killing effects. Approaches such as HSF1 gene-knockdown and small molecule inhibitors like KRIBB11 are explored to downregulate the HSR and augment tumor destruction. We emphasize the impact of HSR inhibition on cancer cell viability, mEHT sensitivity, and potential synergistic effects, addressing challenges and future directions. This understanding offers opportunities for optimizing treatment strategies and advancing precision medicine in cancer therapy.
Collapse
Affiliation(s)
- Pedro Viana
- Institute of Translational Medicine, Semmelweis University, Tűzoltó utca 37-49, 1094 Budapest, Hungary.
| | - Péter Hamar
- Institute of Translational Medicine, Semmelweis University, Tűzoltó utca 37-49, 1094 Budapest, Hungary.
| |
Collapse
|
13
|
Somu P, Mohanty S, Basavegowda N, Yadav AK, Paul S, Baek KH. The Interplay between Heat Shock Proteins and Cancer Pathogenesis: A Novel Strategy for Cancer Therapeutics. Cancers (Basel) 2024; 16:638. [PMID: 38339390 PMCID: PMC10854888 DOI: 10.3390/cancers16030638] [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: 01/15/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Heat shock proteins (HSPs) are developmentally conserved families of protein found in both prokaryotic and eukaryotic organisms. HSPs are engaged in a diverse range of physiological processes, including molecular chaperone activity to assist the initial protein folding or promote the unfolding and refolding of misfolded intermediates to acquire the normal or native conformation and its translocation and prevent protein aggregation as well as in immunity, apoptosis, and autophagy. These molecular chaperonins are classified into various families according to their molecular size or weight, encompassing small HSPs (e.g., HSP10 and HSP27), HSP40, HSP60, HSP70, HSP90, and the category of large HSPs that include HSP100 and ClpB proteins. The overexpression of HSPs is induced to counteract cell stress at elevated levels in a variety of solid tumors, including anticancer chemotherapy, and is closely related to a worse prognosis and therapeutic resistance to cancer cells. HSPs are also involved in anti-apoptotic properties and are associated with processes of cancer progression and development, such as metastasis, invasion, and cell proliferation. This review outlines the previously mentioned HSPs and their significant involvement in diverse mechanisms of tumor advancement and metastasis, as well as their contribution to identifying potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Prathap Somu
- Department of Biotechnology and Chemical Engineering, School of Civil & Chemical Engineering, Manipal University Jaipur, Dehmi Kalan, Jaipur 303007, India;
| | - Sonali Mohanty
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, India;
| | - Nagaraj Basavegowda
- Department of Biotechnology, Yeungnam University, Gyeongsan 38451, Republic of Korea;
| | - Akhilesh Kumar Yadav
- Department of Environmental Engineering and Management, Chaoyang University of Technology, Taichung 413310, Taiwan;
- Department of Bioengineering, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, India
| | - Subhankar Paul
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, India;
| | - Kwang-Hyun Baek
- Department of Biotechnology, Yeungnam University, Gyeongsan 38451, Republic of Korea;
| |
Collapse
|
14
|
Simanjuntak MV, Jauhar MM, Syaifie PH, Arda AG, Mardliyati E, Shalannanda W, Hermanto BR, Anshori I. Revealing Propolis Potential Activity on Inhibiting Estrogen Receptor and Heat Shock Protein 90 Overexpressed in Breast Cancer by Bioinformatics Approaches. Bioinform Biol Insights 2024; 18:11779322231224187. [PMID: 38274992 PMCID: PMC10809879 DOI: 10.1177/11779322231224187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Breast cancer is the most commonly diagnosed cancer globally, with the highest incidence of breast cancer occurring in Asian countries including Indonesia. Among the types of breast cancer, the estrogen receptor (ER)-positive subtype which is prominent with estrogen receptor alpha (ERα) and heat shock protein 90 (HSP90) overexpression genes becomes the most prevalent than the others, approximately 75% of all breast cancer cases. ERα and HSP90 play a role in breast cancer activities including breast tumor growth, invasion, and metastasis mechanism. Propolis, a natural bee product, has been explored for its anticancer activity. However, there is lack of studies that evaluated the potential inhibitor from propolis compounds to the ERα and HSP90 proteins. Therefore, this article focuses on examining the correlation between ERα and HSP90's role in breast cancer and investigating the potential of 93 unique propolis compositions in inhibiting these genes in breast cancer using in silico approaches. This study revealed the positive correlation between ERα and HSP90 genes in breast cancer disease development. Furthermore, we also found novel potential bioactive compounds of propolis against breast cancer through binding with ERα and HSP90; they were 3',4',7-trihydroxyisoflavone and baicalein-7-O-β-D glucopyranoside, respectively. Further research on these compounds is needed to elucidate deeper mechanisms and activity in the real biological system to develop new breast cancer drug treatments.
Collapse
Affiliation(s)
- Masriana Vivi Simanjuntak
- Biomedical Engineering Department, School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, Indonesia
| | - Muhammad Miftah Jauhar
- Center of Excellences Life Sciences, Nano Center Indonesia, South Tangerang, Indonesia
- Biomedical Engineering, The Graduate School of Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Putri Hawa Syaifie
- Center of Excellences Life Sciences, Nano Center Indonesia, South Tangerang, Indonesia
| | - Adzani Gaisani Arda
- Center of Excellences Life Sciences, Nano Center Indonesia, South Tangerang, Indonesia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Etik Mardliyati
- Research Center for Vaccine and Drug, National Research and Innovation Agency (BRIN), Cibinong, Indonesia
| | - Wervyan Shalannanda
- Biomedical Engineering Department, School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, Indonesia
| | - Beni Rio Hermanto
- Biomedical Engineering Department, School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, Indonesia
| | - Isa Anshori
- Biomedical Engineering Department, School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, Indonesia
| |
Collapse
|
15
|
Dunn KA, MacDonald E, MacDonald T, Kulkarni K. Bacterial heat shock protein genes during induction chemotherapy in pediatric patients with acute lymphoblastic leukemia. Future Oncol 2024; 20:17-23. [PMID: 38189148 DOI: 10.2217/fon-2023-0263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024] Open
Abstract
Background: Heat shock proteins (HSP) protect cancer cells. Gastrointestinal bacteria contain HSP genes and can release extracellular vesicles which act as biological shuttles. Stress from treatment may result in a microbial community with more HSP genes, which could contribute to circulating HSP levels. Methods: The authors examined the abundance of five bacterial HSP genes pre-treatment and during induction in stool sequences from 30 pediatric acute lymphoblastic leukemia patients. Results: Decreased mean HTPG counts (p = 0.0024) pre-treatment versus induction were observed. During induction, HTPG, Shannon diversity and Bacteroidetes decreased (p = 7.5e-4; 1.1e-3; 8.6e-4), while DNAK and Firmicutes increased (p = 6.9e-3; 9.2e-4). Conclusion: Understanding microbial HSP gene community changes with treatment is the first step in determining if bacterial HSPs are important to the tumor microenvironment and leukemia treatment.
Collapse
Affiliation(s)
- Katherine A Dunn
- Department of Pediatrics, Division of Hematology Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
- Department of Biology, Dalhousie University, Halifax, NS, Canada
- Institute for Comparative Genomics, Dalhousie University, Halifax, NS, Canada
| | - Emma MacDonald
- Department of Pediatrics, Division of Hematology Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
| | - Tamara MacDonald
- Department of Pharmacy, IWK Health, Halifax, NS, Canada
- Faculty of Health Professions, Dalhousie University, Halifax, NS, Canada
| | - Ketan Kulkarni
- Department of Pediatrics, Division of Hematology Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
| |
Collapse
|
16
|
Torres OKMR, da Silva LAB, Freitas RDA, da Silveira ÉJD, Miguel MCDC. Immunoexpression of HSP27 does not seem to influence the prognosis of oral tongue squamous cell carcinoma. Braz Dent J 2023; 34:125-133. [PMID: 38133467 PMCID: PMC10759947 DOI: 10.1590/0103-6440202305036] [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: 05/05/2022] [Accepted: 08/22/2023] [Indexed: 12/23/2023] Open
Abstract
New methods of early detection and risk assessment have been studied aiming to predict the prognosis of patients and directing a specialized treatment of the oral tongue squamous cell carcinoma (OTSCC). In this context, several molecular biomarkers have been investigated for this purpose, and, among them, the heat shock protein 27 (HSP27) can be named. The study aimed to analyze whether heat shock protein 27 (HSP27) exerts any influence on OTSCC, correlating its immunoexpression with clinicopathological parameters, and patient survival. The sample comprised 55 OTSCC cases and 20 normal oral mucosa specimens. The malignancy grading systems proposed by the WHO in 2005, Brandwein-Gensler et al., and Almangush et al. were applied in a histomorphological study. HSP27 expressions were evaluated through the Immunoreactivity Score System (IRS). Significant values were considered at p <0.05 for all statistical tests. Higher IRS results were observed for normal oral mucosa specimens when compared to OTSCC cases (p <0.001). No significant associations between HSP27 immunostaining, the analyzed clinicopathological parameters and patient survival were observed. The results of the present study indicate lower HSP27 expression in OTSCC cases compared to normal oral mucosa specimens. Thus, HSP27 expression does not seem to influence patient prognosis.
Collapse
Affiliation(s)
- Ondina Karla Mousinho Rocha Torres
- Post graduate Program in Dental Sciences, Department of Oral Pathology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Luiz Arthur Barbosa da Silva
- Post graduate Program in Dental Sciences, Department of Oral Pathology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Roseana de Almeida Freitas
- Department of Oral Pathology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | | | | |
Collapse
|
17
|
Zhou W, Zeng W, Zheng D, Yang X, Qing Y, Zhou C, Liu X. Construction of a prognostic model for lung adenocarcinoma based on heat shock protein-related genes and immune analysis. Cell Stress Chaperones 2023; 28:821-834. [PMID: 37691069 PMCID: PMC10746678 DOI: 10.1007/s12192-023-01374-5] [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: 06/01/2023] [Revised: 08/07/2023] [Accepted: 08/20/2023] [Indexed: 09/12/2023] Open
Abstract
Lung adenocarcinoma (LUAD) represents a prevalent form of cancer, with low early diagnosis rates and high mortality rates, posing a global health challenge. Heat shock proteins (HSPs) assume a crucial role within the tumor immune microenvironment (TME) of LUAD. Here, a collection of 97 HSP-related genes (HSPGs) was assembled based on prior literature reports, of which 36 HSPGs were differentially expressed in LUAD. In The Cancer Genome Atlas (TCGA) cohort, we constructed a prognostic model for risk stratification and prognosis prediction by integrating 13 HSPGs. In addition, the prognostic significance and predictive efficacy of the HSP-related riskscore were examined and validated in the Gene Expression Omnibus (GEO) cohort. To facilitate the clinical use of this riskscore, we also established a nomogram scale by verifying its effectiveness through different methods. In light of these outcomes, we concluded a significant correlation between HSPs and TME in LUAD, and the riskscore can be a reliable prognostic indicator. Furthermore, this study evaluated the differences in immunophenoscore, tumor immune dysfunction and exclusion score, and sensitivity to several common chemotherapy drugs among LUAD individuals in different risk groups, which may aid in clinical decision-making for immune therapy and chemotherapy in LUAD individuals.
Collapse
Affiliation(s)
- Wangyan Zhou
- Department of Medical Record, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang City, 421001, Hunan Province, China
| | - Wei Zeng
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Jiefang Avenue 35, Hengyang City, 421001, Hunan Province, China
| | - Dayang Zheng
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Jiefang Avenue 35, Hengyang City, 421001, Hunan Province, China
| | - Xu Yang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Jiefang Avenue 35, Hengyang City, 421001, Hunan Province, China
| | - Yongcheng Qing
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Jiefang Avenue 35, Hengyang City, 421001, Hunan Province, China
| | - Chunxiang Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Jiefang Avenue 35, Hengyang City, 421001, Hunan Province, China
| | - Xiang Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Jiefang Avenue 35, Hengyang City, 421001, Hunan Province, China.
| |
Collapse
|
18
|
Boyd RA, Majumder S, Stiban J, Mavodza G, Straus AJ, Kempelingaiah SK, Reddy V, Hannun YA, Obeid LM, Senkal CE. The heat shock protein Hsp27 controls mitochondrial function by modulating ceramide generation. Cell Rep 2023; 42:113081. [PMID: 37689067 PMCID: PMC10591768 DOI: 10.1016/j.celrep.2023.113081] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/24/2023] [Accepted: 08/18/2023] [Indexed: 09/11/2023] Open
Abstract
Sphingolipids have key functions in membrane structure and cellular signaling. Ceramide is the central molecule of the sphingolipid metabolism and is generated by ceramide synthases (CerS) in the de novo pathway. Despite their critical function, mechanisms regulating CerS remain largely unknown. Using an unbiased proteomics approach, we find that the small heat shock protein 27 (Hsp27) interacts specifically with CerS1 but not other CerS. Functionally, our data show that Hsp27 acts as an endogenous inhibitor of CerS1. Wild-type Hsp27, but not a mutant deficient in CerS1 binding, inhibits CerS1 activity. Additionally, silencing of Hsp27 enhances CerS1-generated ceramide accumulation in cells. Moreover, phosphorylation of Hsp27 modulates Hsp27-CerS1 interaction and CerS1 activity in acute stress-response conditions. Biologically, we show that Hsp27 knockdown impedes mitochondrial function and induces lethal mitophagy in a CerS1-dependent manner. Overall, we identify an important mode of CerS1 regulation and CerS1-mediated mitophagy through protein-protein interaction with Hsp27.
Collapse
Affiliation(s)
- Rowan A Boyd
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA
| | - Saurav Majumder
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA
| | - Johnny Stiban
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA; Department of Biology and Biochemistry, Birzeit University, Ramallah, Palestine
| | - Grace Mavodza
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA
| | - Alexandra J Straus
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA
| | - Sachin K Kempelingaiah
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA
| | - Varun Reddy
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yusuf A Hannun
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Lina M Obeid
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA; Northport Veterans Affairs Medical Center, Northport, NY 11768, USA
| | - Can E Senkal
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23398, USA.
| |
Collapse
|
19
|
Amiri M, Molavi O, Sabetkam S, Jafari S, Montazersaheb S. Stimulators of immunogenic cell death for cancer therapy: focusing on natural compounds. Cancer Cell Int 2023; 23:200. [PMID: 37705051 PMCID: PMC10500939 DOI: 10.1186/s12935-023-03058-7] [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: 04/05/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023] Open
Abstract
A growing body of evidence indicates that the anticancer effect of the immune system can be activated by the immunogenic modulation of dying cancer cells. Cancer cell death, as a result of the activation of an immunomodulatory response, is called immunogenic cell death (ICD). This regulated cell death occurs because of increased immunogenicity of cancer cells undergoing ICD. ICD plays a crucial role in stimulating immune system activity in cancer therapy. ICD can therefore be an innovative route to improve anticancer immune responses associated with releasing damage-associated molecular patterns (DAMPs). Several conventional and chemotherapeutics, as well as preclinically investigated compounds from natural sources, possess immunostimulatory properties by ICD induction. Natural compounds have gained much interest in cancer therapy owing to their low toxicity, low cost, and inhibiting cancer cells by interfering with different mechanisms, which are critical in cancer progression. Therefore, identifying natural compounds with ICD-inducing potency presents agents with promising potential in cancer immunotherapy. Naturally derived compounds are believed to act as immunoadjuvants because they elicit cancer stress responses and DAMPs. Acute exposure to DAMP molecules can activate antigen-presenting cells (APCs), such as dendritic cells (DCs), which leads to downstream events by cytotoxic T lymphocytes (CTLs) and natural killer cells (NKs). Natural compounds as inducers of ICD may be an interesting approach to ICD induction; however, parameters that determine whether a compound can be used as an ICD inducer should be elucidated. Here, we aimed to discuss the impact of multiple ICD inducers, mainly focusing on natural agents, including plant-derived, marine molecules, and bacterial-based compounds, on the release of DAMP molecules and the activation of the corresponding signaling cascades triggering immune responses. In addition, the potential of synthetic agents for triggering ICD is also discussed.
Collapse
Affiliation(s)
- Mina Amiri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahnaz Sabetkam
- Department of Anatomy, Faculty of Medicine, university of Kyrenia, Kyrenia, Northern Cyprus
- Department of Anatomy and histopathology, Faculty of medicine, Tabriz medical sciences, Islamic Azad University, Tabriz, Iran
| | - Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
20
|
Mazurakova A, Solarova Z, Koklesova L, Caprnda M, Prosecky R, Khakymov A, Baranenko D, Kubatka P, Mirossay L, Kruzliak P, Solar P. Heat shock proteins in cancer - Known but always being rediscovered: Their perspectives in cancer immunotherapy. Adv Med Sci 2023; 68:464-473. [PMID: 37926002 DOI: 10.1016/j.advms.2023.10.005] [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: 12/27/2022] [Revised: 02/14/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023]
Abstract
Heat shock proteins (HSPs) represent cellular chaperones that are classified into several families, including HSP27, HSP40, HSP60, HSP70, and HSP90. The role of HSPs in the cell includes the facilitation of protein folding and maintaining protein structure. Both processes play crucial roles during stress conditions in the cell such as heat shock, degradation, and hypoxia. Moreover, HSPs are important modulators of cellular proliferation and differentiation, and are strongly associated with the molecular orchestration of carcinogenesis. The expression and/or activity of HSPs in cancer cells is generally abnormally high and is associated with increased metastatic potential and activity of cancer stem cells, more pronounced angiogenesis, downregulated apoptosis, and the resistance to anticancer therapy in many patients. Based on the mentioned reasons, HSPs have strong potential as valid diagnostic, prognostic, and therapeutic biomarkers in clinical oncology. In addition, numerous papers describe the role of HSPs as chaperones in the regulation of immune responses inside and outside the cell. Importantly, highly expressed/activated HSPs may be inhibited via immunotherapeutic targets in various types of cancers. The aim of this work is to provide a comprehensive overview of the relationship between HSPs and the tumor cell with the intention of highlighting the potential use of HSPs in personalized cancer management.
Collapse
Affiliation(s)
- Alena Mazurakova
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Zuzana Solarova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | - Robert Prosecky
- 2nd Department of Internal Medicine, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic; International Clinical Research Centre, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Artur Khakymov
- International Research Centre "Biotechnologies of the Third Millennium", Faculty of Biotechnologies (BioTech), ITMO University, Saint-Petersburg, Russian Federation
| | - Denis Baranenko
- International Research Centre "Biotechnologies of the Third Millennium", Faculty of Biotechnologies (BioTech), ITMO University, Saint-Petersburg, Russian Federation
| | - Peter Kubatka
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovakia
| | - Peter Kruzliak
- 2nd Department of Surgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic.
| | - Peter Solar
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovakia.
| |
Collapse
|
21
|
Magyar CTJ, Vashist YK, Stroka D, Kim-Fuchs C, Berger MD, Banz VM. Heat shock protein 90 (HSP90) inhibitors in gastrointestinal cancer: where do we currently stand?-A systematic review. J Cancer Res Clin Oncol 2023; 149:8039-8050. [PMID: 36966394 PMCID: PMC10374781 DOI: 10.1007/s00432-023-04689-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/09/2023] [Indexed: 03/27/2023]
Abstract
PURPOSE Dysregulated expression of heat shock proteins (HSP) plays a fundamental role in tumor development and progression. Consequently, HSP90 may be an effective tumor target in oncology, including the treatment of gastrointestinal cancers. METHODS We carried out a systematic review of data extracted from clinicaltrials.gov and pubmed.gov, which included all studies available until January 1st, 2022. The published data was evaluated using primary and secondary endpoints, particularly with focus on overall survival, progression-free survival, and rate of stable disease. RESULTS Twenty trials used HSP90 inhibitors in GI cancers, ranging from phase I to III clinical trials. Most studies assessed HSP90 inhibitors as a second line treatment. Seventeen of the 20 studies were performed prior to 2015 and only few studies have results pending. Several studies were terminated prematurely, due to insufficient efficacy or toxicity. Thus far, the data suggests that HSP90 inhibitor NVP-AUY922 might improve outcome for colorectal cancer and gastrointestinal stromal tumors. CONCLUSION It currently remains unclear which subgroup of patients might benefit from HSP90 inhibitors and at what time point these inhibitors may be beneficial. There are only few new or ongoing studies initiated during the last decade.
Collapse
Affiliation(s)
- Christian Tibor Josef Magyar
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | | | - Deborah Stroka
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Corina Kim-Fuchs
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Martin D. Berger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Vanessa M. Banz
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
22
|
Chinnappan R, Mir TA, Alsalameh S, Makhzoum T, Alzhrani A, Alnajjar K, Adeeb S, Al Eman N, Ahmed Z, Shakir I, Al-Kattan K, Yaqinuddin A. Emerging Biosensing Methods to Monitor Lung Cancer Biomarkers in Biological Samples: A Comprehensive Review. Cancers (Basel) 2023; 15:3414. [PMID: 37444523 DOI: 10.3390/cancers15133414] [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: 05/21/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Lung cancer is the most commonly diagnosed of all cancers and one of the leading causes of cancer deaths among men and women worldwide, causing 1.5 million deaths every year. Despite developments in cancer treatment technologies and new pharmaceutical products, high mortality and morbidity remain major challenges for researchers. More than 75% of lung cancer patients are diagnosed in advanced stages, leading to poor prognosis. Lung cancer is a multistep process associated with genetic and epigenetic abnormalities. Rapid, accurate, precise, and reliable detection of lung cancer biomarkers in biological fluids is essential for risk assessment for a given individual and mortality reduction. Traditional diagnostic tools are not sensitive enough to detect and diagnose lung cancer in the early stages. Therefore, the development of novel bioanalytical methods for early-stage screening and diagnosis is extremely important. Recently, biosensors have gained tremendous attention as an alternative to conventional methods because of their robustness, high sensitivity, inexpensiveness, and easy handling and deployment in point-of-care testing. This review provides an overview of the conventional methods currently used for lung cancer screening, classification, diagnosis, and prognosis, providing updates on research and developments in biosensor technology for the detection of lung cancer biomarkers in biological samples. Finally, it comments on recent advances and potential future challenges in the field of biosensors in the context of lung cancer diagnosis and point-of-care applications.
Collapse
Affiliation(s)
- Raja Chinnappan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, Transplant Research & Innovation Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Tanveer Ahmad Mir
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, Transplant Research & Innovation Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | | | - Tariq Makhzoum
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Alaa Alzhrani
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, Transplant Research & Innovation Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Khalid Alnajjar
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Salma Adeeb
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Noor Al Eman
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Zara Ahmed
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Ismail Shakir
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Khaled Al-Kattan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
23
|
Alessandrini L, Astolfi L, Daloiso A, Sbaraglia M, Mondello T, Zanoletti E, Franz L, Marioni G. Diagnostic, Prognostic, and Therapeutic Role for Angiogenesis Markers in Head and Neck Squamous Cell Carcinoma: A Narrative Review. Int J Mol Sci 2023; 24:10733. [PMID: 37445908 DOI: 10.3390/ijms241310733] [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: 04/23/2023] [Revised: 06/14/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Despite refinements to diagnostic and therapeutic approaches over the last two decades, the outcome of patients with head and neck squamous cell carcinoma (HNSCC) has not shown substantial improvements, especially regarding those with advanced-stage disease. Angiogenesis is believed to be a turning point in the development of solid tumors, being a premise for mass growth and potential distant dissemination. Cancer-induced angiogenesis is a result of increased expression of angiogenic factors, decreased expression of anti-angiogenic factors, or a combination of both. The assessment of angiogenesis has also emerged as a potentially useful biological prognostic and predictive factor in HNSCC. The aim of this review is to assess the level of current knowledge on the neo-angiogenesis markers involved in the biology, behavior, and prognosis of HNSCC. A search (between 1 January 2012 and 10 October 2022) was run in PubMed, Scopus, and Web of Science electronic databases. After full-text screening and application of inclusion/exclusion criteria, 84 articles are included. The current knowledge and debate on angiogenesis in HNSCC presented in the eligible articles are stratified as follows: (i) diagnostic markers; (ii) prognostic markers; (iii) predictive markers; and (iv) markers with a potential therapeutic role. Angiogenesis is a biological and pathological indicator of malignancies progression and has negative implications in prognosis of some solid tumors; several signals capable of tripping the "angiogenic switch" have also been identified in HNSCC. Although several studies suggested that antiangiogenic agents might be a valuable adjunct to conventional chemo-radiation of HNSCC, their long-term therapeutic value remains uncertain. Further investigations are required on combinations of antiangiogenic agents with conventional chemotherapeutic ones, immunotherapeutic and molecularly targeted agents in HNSCC. Additional data are necessary to pinpoint which patients could benefit most from these treatments.
Collapse
Affiliation(s)
- Lara Alessandrini
- Surgical Pathology and Cytopathology Unit, Department of Medicine (DIMED), University of Padova, 35100 Padova, Italy
| | - Laura Astolfi
- Bioacustic Research Laboratory, Department of Neuroscience (DNS), University of Padova, 35100 Padova, Italy
| | - Antonio Daloiso
- Otolaryngology Section, Department of Neuroscience (DNS), University of Padova, 35100 Padova, Italy
| | - Marta Sbaraglia
- Surgical Pathology and Cytopathology Unit, Department of Medicine (DIMED), University of Padova, 35100 Padova, Italy
| | - Tiziana Mondello
- Otolaryngology Section, Department of Neuroscience (DNS), University of Padova, 35100 Padova, Italy
| | - Elisabetta Zanoletti
- Otolaryngology Section, Department of Neuroscience (DNS), University of Padova, 35100 Padova, Italy
| | - Leonardo Franz
- Otolaryngology Section, Department of Neuroscience (DNS), University of Padova, 35100 Padova, Italy
- Phoniatrics and Audiology Unit, Department of Neuroscience (DNS), University of Padova, 31100 Treviso, Italy
- Artificial Intelligence in Medicine and Innovation in Clinical Research and Methodology (PhD Program), Department of Clinical and Experimental Sciences, University of Brescia, 25100 Brescia, Italy
| | - Gino Marioni
- Phoniatrics and Audiology Unit, Department of Neuroscience (DNS), University of Padova, 31100 Treviso, Italy
| |
Collapse
|
24
|
Zou Y, Shi H, Liu N, Wang H, Song X, Liu B. Mechanistic insights into heat shock protein 27, a potential therapeutic target for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1195464. [PMID: 37252119 PMCID: PMC10219228 DOI: 10.3389/fcvm.2023.1195464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
Heat shock protein 27 (HSP27) is a small chaperone protein that is overexpressed in a variety of cellular stress states. It is involved in regulating proteostasis and protecting cells from multiple sources of stress injury by stabilizing protein conformation and promoting the refolding of misfolded proteins. Previous studies have confirmed that HSP27 is involved in the development of cardiovascular diseases and plays an important regulatory role in this process. Herein, we comprehensively and systematically summarize the involvement of HSP27 and its phosphorylated form in pathophysiological processes, including oxidative stress, inflammatory responses, and apoptosis, and further explore the potential mechanisms and possible roles of HSP27 in the diagnosis and treatment of cardiovascular diseases. Targeting HSP27 is a promising future strategy for the treatment of cardiovascular diseases.
Collapse
|
25
|
Geitgey DK, Lee M, Cottrill KA, Jaffe M, Pilcher W, Bhasin S, Randall J, Ross AJ, Salemi M, Castillo-Castrejon M, Kilgore MB, Brown AC, Boss JM, Johnston R, Fitzpatrick AM, Kemp ML, English R, Weaver E, Bagchi P, Walsh R, Scharer CD, Bhasin M, Chandler JD, Haynes KA, Wellberg EA, Henry CJ. The 'omics of obesity in B-cell acute lymphoblastic leukemia. J Natl Cancer Inst Monogr 2023; 2023:12-29. [PMID: 37139973 PMCID: PMC10157791 DOI: 10.1093/jncimonographs/lgad014] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/12/2023] [Accepted: 03/17/2023] [Indexed: 05/05/2023] Open
Abstract
The obesity pandemic currently affects more than 70 million Americans and more than 650 million individuals worldwide. In addition to increasing susceptibility to pathogenic infections (eg, SARS-CoV-2), obesity promotes the development of many cancer subtypes and increases mortality rates in most cases. We and others have demonstrated that, in the context of B-cell acute lymphoblastic leukemia (B-ALL), adipocytes promote multidrug chemoresistance. Furthermore, others have demonstrated that B-ALL cells exposed to the adipocyte secretome alter their metabolic states to circumvent chemotherapy-mediated cytotoxicity. To better understand how adipocytes impact the function of human B-ALL cells, we used a multi-omic RNA-sequencing (single-cell and bulk transcriptomic) and mass spectroscopy (metabolomic and proteomic) approaches to define adipocyte-induced changes in normal and malignant B cells. These analyses revealed that the adipocyte secretome directly modulates programs in human B-ALL cells associated with metabolism, protection from oxidative stress, increased survival, B-cell development, and drivers of chemoresistance. Single-cell RNA sequencing analysis of mice on low- and high-fat diets revealed that obesity suppresses an immunologically active B-cell subpopulation and that the loss of this transcriptomic signature in patients with B-ALL is associated with poor survival outcomes. Analyses of sera and plasma samples from healthy donors and those with B-ALL revealed that obesity is associated with higher circulating levels of immunoglobulin-associated proteins, which support observations in obese mice of altered immunological homeostasis. In all, our multi-omics approach increases our understanding of pathways that may promote chemoresistance in human B-ALL and highlight a novel B-cell-specific signature in patients associated with survival outcomes.
Collapse
Affiliation(s)
- Delaney K Geitgey
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
| | - Miyoung Lee
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
| | - Kirsten A Cottrill
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Maya Jaffe
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - William Pilcher
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Swati Bhasin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
- Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Jessica Randall
- Emory Integrated Computational Core, Emory University, Atlanta, GA, USA
| | - Anthony J Ross
- Riley Children’s Health, Indiana University Health, Indianapolis, IN, USA
| | - Michelle Salemi
- Proteomics Core Facility, University of California Davis Genome Center, Davis, 95616, CA
| | - Marisol Castillo-Castrejon
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew B Kilgore
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ayjha C Brown
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Rich Johnston
- Emory Integrated Computational Core, Emory University, Atlanta, GA, USA
| | - Anne M Fitzpatrick
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Melissa L Kemp
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Emory Integrated Proteomics Core, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Eric Weaver
- Shimadzu Scientific Instruments, Columbia, MD, USA
| | - Pritha Bagchi
- Emory Integrated Proteomics Core, Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan Walsh
- Shimadzu Scientific Instruments, Columbia, MD, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Manoj Bhasin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Children’s Healthcare of Atlanta, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Joshua D Chandler
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Karmella A Haynes
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Elizabeth A Wellberg
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Curtis J Henry
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
- Children’s Healthcare of Atlanta, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| |
Collapse
|
26
|
Skowron MA, Kotthoff M, Bremmer F, Ruhnke K, Parmaksiz F, Richter A, Küffer S, Reuter-Jessen K, Pauls S, Stefanski A, Ströbel P, Stühler K, Nettersheim D. Targeting CLDN6 in germ cell tumors by an antibody-drug-conjugate and studying therapy resistance of yolk-sac tumors to identify and screen specific therapeutic options. Mol Med 2023; 29:40. [PMID: 36991316 PMCID: PMC10053054 DOI: 10.1186/s10020-023-00636-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Being the standard-of-care for four decades, cisplatin-based chemotherapy is highly efficient in treating germ cell tumors (GCT). However, often refractory patients present with a remaining (resistant) yolk-sac tumor (YST(-R)) component, resulting in poor prognosis due to lack of novel treatment options besides chemotherapy and surgery. The aim of this study was to identify novel targets for the treatment of YST by deciphering the molecular mechanisms of therapy resistance. Additionally, we screened the cytotoxic efficacy of a novel antibody-drug-conjugate targeting CLDN6 (CLDN6-ADC), as well as pharmacological inhibitors to target specifically YST. METHODS Protein and mRNA levels of putative targets were measured by flow cytometry, immunohistochemical stainings, mass spectrometry of formalin-fixed paraffin-embedded tissues, phospho-kinase arrays, or qRT-PCR. Cell viability, apoptosis and cell cycle assays of GCT and non-cancerous cells were performed using XTT cell viability assays or Annexin V / propidium iodide flow cytometry, respectively. Druggable genomic alterations of YST(-R) tissues were identified by the TrueSight Oncology 500 assay. RESULTS We demonstrated that treatment with a CLDN6-ADC enhanced apoptosis induction specifically in CLDN6+ GCT cells in comparison with non-cancerous controls. In a cell line-dependent manner, either an accumulation in the G2 / M cell cycle phase or a mitotic catastrophe was observed. Based on mutational and proteome profiling, this study identified drugs targeting the FGF, VGF, PDGF, mTOR, CHEK1, AURKA, or PARP signaling pathways as promising approaches to target YST. Further, we identified factors relevant for MAPK signaling, translational initiation and RNA binding, extracellular matrix-related processes as well as oxidative stress and immune response to be involved in therapy resistance. CONCLUSIONS In summary, this study offers a novel CLDN6-ADC to target GCT. Additionally, this study presents novel pharmacological inhibitors blocking FGF, VGF, PDGF, mTOR, CHEK1, AURKA, or PARP signaling for the treatment of (refractory) YST patients. Finally, this study shed light on the mechanisms of therapy resistance in YST.
Collapse
Affiliation(s)
- Margaretha A Skowron
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Mara Kotthoff
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Felix Bremmer
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Katja Ruhnke
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Fatma Parmaksiz
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Annika Richter
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Küffer
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Stella Pauls
- Molecular Proteomics Laboratory, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Anja Stefanski
- Molecular Proteomics Laboratory, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Nettersheim
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
27
|
Nitzsche B, Höpfner M, Biersack B. Synthetic Small Molecule Modulators of Hsp70 and Hsp40 Chaperones as Promising Anticancer Agents. Int J Mol Sci 2023; 24:4083. [PMID: 36835501 PMCID: PMC9964478 DOI: 10.3390/ijms24044083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/22/2023] Open
Abstract
A class of chaperones dubbed heat shock protein 70 (Hsp70) possesses high relevance in cancer diseases due to its cooperative activity with the well-established anticancer target Hsp90. However, Hsp70 is closely connected with a smaller heat shock protein, Hsp40, forming a formidable Hsp70-Hsp40 axis in various cancers, which serves as a suitable target for anticancer drug design. This review summarizes the current state and the recent developments in the field of (semi-)synthetic small molecule inhibitors directed against Hsp70 and Hsp40. The medicinal chemistry and anticancer potential of pertinent inhibitors are discussed. Since Hsp90 inhibitors have entered clinical trials but have exhibited severe adverse effects and drug resistance formation, potent Hsp70 and Hsp40 inhibitors may play a significant role in overcoming the drawbacks of Hsp90 inhibitors and other approved anticancer drugs.
Collapse
Affiliation(s)
- Bianca Nitzsche
- Institute for Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Höpfner
- Institute for Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Bernhard Biersack
- Organische Chemie 1, Universität Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| |
Collapse
|
28
|
Gonnella R, Arena A, Zarrella R, Gilardini Montani MS, Santarelli R, Cirone M. HSPs/STAT3 Interplay Sustains DDR and Promotes Cytokine Release by Primary Effusion Lymphoma Cells. Int J Mol Sci 2023; 24:ijms24043933. [PMID: 36835344 PMCID: PMC9959463 DOI: 10.3390/ijms24043933] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Primary effusion lymphoma (PEL) is a rare and aggressive B-cell lymphoma, against which current therapies usually fail. In the present study, we show that targeting HSPs, such as HSP27, HSP70 and HSP90, could be an efficient strategy to reduce PEL cell survival, as it induces strong DNA damage, which correlated with an impairment of DDR. Moreover, as HSP27, HSP70 and HSP90 cross talk with STAT3, their inhibition results in STAT3 de-phosphorylation and. On the other hand, the inhibition of STAT3 may downregulate these HSPs. These findings suggest that targeting HSPs has important implications in cancer therapy, as it can reduce the release of cytokines by PEL cells, which, besides affecting their own survival, could negatively influence anti-cancer immune response.
Collapse
|
29
|
Sasaya T, Kubo T, Murata K, Mizue Y, Sasaki K, Yanagawa J, Imagawa M, Kato H, Tsukahara T, Kanaseki T, Tamura Y, Miyazaki A, Hirohashi Y, Torigoe T. Cisplatin-induced HSF1-HSP90 axis enhances the expression of functional PD-L1 in oral squamous cell carcinoma. Cancer Med 2023; 12:4605-4615. [PMID: 36200687 PMCID: PMC9972142 DOI: 10.1002/cam4.5310] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/07/2022] [Accepted: 09/20/2022] [Indexed: 11/09/2022] Open
Abstract
Immune checkpoint inhibitor-based cancer immunotherapy has provided an additional therapeutic option for oral squamous cell carcinoma (OSCC) with recurrence or distant metastases. However, further improvement of OSCC treatment is required to develop the optimal combination or order for chemoradiotherapy and immunotherapy. Along with the accumulation of clinical knowledge and evidence, it is also essential to clarify the biological impact of chemo-radiotherapeutic agents on the cancer immune microenvironment. In this study, we investigated the effects of cisplatin (CDDP), a key therapeutic agent for OSCC, on programmed death-ligand 1 (PD-L1) expression in OSCC lines. Although CDDP treatment increased the surface levels of PD-L1 on OSCC cell lines, the gene and total protein expression levels of PD-L1 were not altered. We also demonstrated that the phosphorylation of heat shock factor 1 and heat shock protein 90 was involved in this process. In addition, CDDP-induced PD-L1 attenuated the target-specific cytotoxic T lymphocyte reaction to OSCC. These results provide an immunobiological basis for the response of OSCC to CDDP and will contribute to our biological understanding of the action of novel combination therapy including immunotherapy together with platinum-based chemotherapy for OSCC.
Collapse
Affiliation(s)
- Takashi Sasaya
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan.,Department of Oral Surgery, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Terufumi Kubo
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Kenji Murata
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Yuka Mizue
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Kenta Sasaki
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Junko Yanagawa
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Makoto Imagawa
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Hirotaka Kato
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan.,Department of Oral Surgery, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Tomohide Tsukahara
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Takayuki Kanaseki
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Yasuaki Tamura
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Akihiro Miyazaki
- Department of Oral Surgery, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Yoshihiko Hirohashi
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University, School of Medicine, Sapporo, Japan
| |
Collapse
|
30
|
Evaluation of the Small Heat Shock Protein Family Members HSPB2 and HSPB3 in Bladder Cancer Prognosis and Progression. Int J Mol Sci 2023; 24:ijms24032609. [PMID: 36768927 PMCID: PMC9917356 DOI: 10.3390/ijms24032609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Bladder cancer (BlCa) represents the sixth most commonly diagnosed type of male malignancy. Due to the clinical heterogeneity of BlCa, novel markers would optimize treatment efficacy and improve prognosis. The small heat shock proteins (sHSP) family is one of the major groups of molecular chaperones responsible for the maintenance of proteome functionality and stability. However, the role of sHSPs in BlCa remains largely unknown. The present study aimed to examine the association between HSPB2 and HSPB3 expression and BlCa progression in patients, and to investigate their role in BlCa cells. For this purpose, a series of experiments including reverse transcription-quantitative PCR, Western blotting, MTT assay and flow cytometry were performed. Initial analyses revealed increased vs. human transitional carcinoma cells, expression levels of the HSPB2 and HSPB3 genes and proteins in high grade BlCa cell lines. Therefore, we then evaluated the clinical significance of the HSPB2 and HSPB3 genes expression levels in bladder tumor samples and matched adjusted normal bladder specimens. Total RNA from 100 bladder tumor samples and 49 paired non-cancerous bladder specimens were isolated, and an accurate SYBR-Green based real-time quantitative polymerase chain reaction (qPCR) protocol was developed to quantify HSPB2 and HSPB3 mRNA levels in the two cohorts of specimens. A significant downregulation of the HSPB2 and HSPB3 genes expression was observed in bladder tumors as compared to matched normal urothelium; yet, increased HSPB2 and HSPB3 levels were noted in muscle-invasive (T2-T4) vs. superficial tumors (TaT1), as well as in high-grade vs. low-grade tumors. Survival analyses highlighted the significantly higher risk for post-treatment disease relapse in TaT1 patients poorly expressing HSPB2 and HSPB3 genes; this effect tended to be inverted in advanced disease stages (muscle-invasive tumors) indicating the biphasic impact of HSPB2, HSPB3 genes in BlCa progression. The pro-survival role of HSPB2 and HSPB3 in advanced tumor cells was also evident by our finding that HSPB2, HSPB3 genes expression silencing in high grade BlCa cells enhanced doxorubicin toxicity. These findings indicate that the HSPB2, HSPB3 chaperone genes have a likely pro-survival role in advanced BlCa; thus, they can be targeted as novel molecular markers to optimize treatment efficacy in BlCa and to limit unnecessary interventions.
Collapse
|
31
|
Kim SA, Lee Y, Ko Y, Kim S, Kim GB, Lee NK, Ahn W, Kim N, Nam GH, Lee EJ, Kim IS. Protein-based nanocages for vaccine development. J Control Release 2023; 353:767-791. [PMID: 36516900 DOI: 10.1016/j.jconrel.2022.12.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Protein nanocages have attracted considerable attention in various fields of nanomedicine due to their intrinsic properties, including biocompatibility, biodegradability, high structural stability, and ease of modification of their surfaces and inner cavities. In vaccine development, these protein nanocages are suited for efficient targeting to and retention in the lymph nodes and can enhance immunogenicity through various mechanisms, including excellent uptake by antigen-presenting cells and crosslinking with multiple B cell receptors. This review highlights the superiority of protein nanocages as antigen delivery carriers based on their physiological and immunological properties such as biodistribution, immunogenicity, stability, and multifunctionality. With a focus on design, we discuss the utilization and efficacy of protein nanocages such as virus-like particles, caged proteins, and artificial caged proteins against cancer and infectious diseases such as coronavirus disease 2019 (COVID-19). In addition, we summarize available knowledge on the protein nanocages that are currently used in clinical trials and provide a general outlook on conventional distribution techniques and hurdles faced, particularly for therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Yeram Lee
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Yeju Ko
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Seohyun Kim
- Department of Research and Development, SHIFTBIO INC., Seoul, Republic of Korea
| | - Gi Beom Kim
- Department of Research and Development, SHIFTBIO INC., Seoul, Republic of Korea
| | - Na Kyeong Lee
- Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Wonkyung Ahn
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Nayeon Kim
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Gi-Hoon Nam
- Department of Research and Development, SHIFTBIO INC., Seoul, Republic of Korea; Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Lee
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea; Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
32
|
Le XT, Lee J, Nguyen NT, Lee WT, Lee ES, Oh KT, Choi HG, Shin BS, Youn YS. Combined phototherapy with metabolic reprogramming-targeted albumin nanoparticles for treating breast cancer. Biomater Sci 2022; 10:7117-7132. [PMID: 36350285 DOI: 10.1039/d2bm01281b] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Triple-negative breast cancer (TNBC) is characterized by rapid tumor growth and resistance to cancer therapy, and has a poor prognosis. Accumulating data have revealed that cancer metabolism relies on both the Warburg effect and oxidative phosphorylation (OXPHOS), which are strongly related to the high proliferation and chemoresistance of cancer cells. Phototherapy is considered as a non-invasive method to precisely control drug activity with reduced side effects. Herein, our group introduced an Abraxane-like nanoplatform, named LCIR NPs, which significantly eradicates cancer cells via synergism between metabolic reprogramming and phototherapy effects. Endowed with mitochondria-targeting residues, the nanoparticles efficiently inhibited mitochondrial complexes I and IV as well as hexokinase II, leading to the depletion of intracellular ATP. Consequently, the photodynamic and photothermal effect triggered by NIR irradiation was enhanced due to the alleviation of hypoxia and the thermoresistance mechanism that rely on mitochondrial metabolism. In vivo experiments showed that the tumor size of mice that received the combination treatment was only 50.7 mm3, which was 21 times smaller than that of the untreated group and was much lower than those of other single treatments after 21 days. Additionally, almost no systemic undesired toxicity was detected during the observation period. We believe that the concept of LCIR as presented here offers a potential platform to overcome the resistance to conventional therapies by the incorporation with the energy metabolism inhibition approach.
Collapse
Affiliation(s)
- Xuan Thien Le
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Junyeong Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Nguyen Thi Nguyen
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Woo Tak Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Eun Seong Lee
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea
| | - Beom Soo Shin
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| |
Collapse
|
33
|
Parma B, Wurdak H, Ceppi P. Harnessing mitochondrial metabolism and drug resistance in non-small cell lung cancer and beyond by blocking heat-shock proteins. Drug Resist Updat 2022; 65:100888. [DOI: 10.1016/j.drup.2022.100888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/10/2022] [Accepted: 10/25/2022] [Indexed: 11/30/2022]
|
34
|
AHSA1 Promotes Proliferation and EMT by Regulating ERK/CALD1 Axis in Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14194600. [PMID: 36230524 PMCID: PMC9562867 DOI: 10.3390/cancers14194600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/13/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Globally, hepatocellular carcinoma is one of the leading causes of cancer-related death. Activator of HSP90 ATPase activity 1(AHSA1) was reported to be involved in regulating the maturation, stability, and degradation of related cancer-promoting proteins. However, the biological function and regulatory mechanism of AHSA1 in hepatocellular carcinoma were unclearly. In this study, we found AHSA1 was upregulated and associated with poor clinical characteristics and prognosis of hepatocellular carcinoma patients. Moreover, AHSA1 promoted proliferation, metastasis, epithelium-mesenchymal transition of hepatocellular carcinoma both in vitro and in vivo by recruiting ERK1/2 and promoting the phosphorylation and inactivation of CALD1. Taken together, our findings provided a mechanistic insight into the role of AHSA1 in hepatocellular carcinoma progression and implied AHSA1 may be a biomarker and therapeutic target for hepatocellular carcinoma patients. Abstract Hepatocellular carcinoma (HCC) is one of the major causes of cancer-related death worldwide. AHSA1 as a chaperone of HSP90 promotes the maturation, stability, and degradation of related cancer-promoting proteins. However, the regulatory mechanism and biological function of AHSA1 in HCC are largely unknown. Actually, we found that AHSA1 was significantly upregulated in HCC tissues and cell lines and was notably correlated with the poor clinical characteristics and prognosis of HCC patients in this study. Furthermore, both in vitro and in vivo, gain- and loss-of-function studies demonstrated that AHSA1 promoted the proliferation, invasion, metastasis, and epithelial-mesenchymal transition (EMT) of HCC. Moreover, the mechanistic study indicated that AHSA1 recruited ERK1/2 and promoted the phosphorylation and inactivation of CALD1, while ERK1/2 phosphorylation inhibitor SCH772984 reversed the role of AHSA1 in the proliferation and EMT of HCC. Furthermore, we demonstrated that the knockdown of CALD1 reversed the inhibition of proliferation and EMT by knocking AHSA1 in HCC. We also illustrated a new molecular mechanism associated with AHSA1 in HCC that was independent of HSP90 and MEK1/2. In summary, AHSA1 may play an oncogenic role in HCC by regulating ERK/CALD1 axis and may serve as a novel therapeutic target for HCC.
Collapse
|
35
|
Javid H, Hashemian P, Yazdani S, Sharbaf Mashhad A, Karimi-Shahri M. The role of heat shock proteins in metastatic colorectal cancer: A review. J Cell Biochem 2022; 123:1704-1735. [PMID: 36063530 DOI: 10.1002/jcb.30326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 01/18/2023]
Abstract
Heat shock proteins (HSPs) are a large molecular chaperone family classified by their molecular weights, including HSP27, HSP40, HSP60, HSP70, HSP90, and HSP110. HSPs are likely to have antiapoptotic properties and participate actively in various processes such as tumor cell proliferation, invasion, metastases, and death. In this review, we discuss comprehensively the functions of HSPs associated with the progression of colorectal cancer (CRC) and metastasis and resistance to cancer therapy. Taken together, HSPs have numerous clinical applications as biomarkers for cancer diagnosis and prognosis and potential therapeutic targets for CRC and its related metastases.
Collapse
Affiliation(s)
- Hossein Javid
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | - Pedram Hashemian
- Jahad Daneshgahi Research Committee, Jahad Daneshgahi Institute, Mashhad, Iran
| | - Shaghayegh Yazdani
- Department of Medical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Laboratory Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - Alireza Sharbaf Mashhad
- Department of Medical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
36
|
Nishimura T, Takadate T, Maeda S, Suzuki T, Minowa T, Fukuda T, Bando Y, Unno M. Disease-related protein co-expression networks are associated with the prognosis of resectable node-positive pancreatic ductal adenocarcinoma. Sci Rep 2022; 12:14709. [PMID: 36038612 PMCID: PMC9424258 DOI: 10.1038/s41598-022-19182-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 08/25/2022] [Indexed: 12/05/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a multifactorial disease, the molecular profile of which remains unclear. This study aimed at unveiling the disease-related protein networks associated with different outcomes of resectable, node-positive PDAC cases. We assessed laser-microdissected cancerous cells from PDAC tissues of a poor outcome group (POG; n = 4) and a better outcome group (BOG; n = 4). Noncancerous pancreatic duct tissues (n = 5) were used as the reference. We identified four representative network modules by applying a weighted network correlation analysis to the obtained quantitative PDAC proteome datasets. Two network modules that were significant for POG were associated with the heat shock response to hypoxia-related stress; in the latter, a large involvement of the non-canonical Hedgehog pathway (regulated by GLI1), the internal ribosome entry site-mediated cap-independent translation, the inositol requiring enzyme 1-alpha (IRE1α)/X-box binding protein 1 pathway of the unfolding protein response (UPR), and the aerobic glycolysis was observed. By contrast, the BOG characteristic module was involved in the inactivation of the UPR pathway via the synoviolin 1-dependent proteasomal degradation of IRE1α, the activation of SOX2, and the loss of PALB2 (partner and localizer of BRCA2) function, all potentially suppressing malignant tumor development. Our findings might facilitate future therapeutic strategies for PDAC.
Collapse
Affiliation(s)
- Toshihide Nishimura
- Department of Translational Medicine Informatics, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan. .,Biosys Technologies, Inc., Tokyo, Tokyo, 153-8904, Japan.
| | - Tatsuyuki Takadate
- Department of Surgery, National Hospital Organization Sendai Medical Center, Sendai, Miyagi, 983-8520, Japan
| | - Shimpei Maeda
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Shimotsuke, Tochigi, 329-0498, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8574, Japan
| | - Takashi Minowa
- Nanotechnology Innovation Station, National Institute for Materials Science, Tsukuba, Ibaraki, Japan
| | - Tetsuya Fukuda
- Biosys Technologies, Inc., Tokyo, Tokyo, 153-8904, Japan
| | - Yasuhiko Bando
- Biosys Technologies, Inc., Tokyo, Tokyo, 153-8904, Japan
| | - Michiaki Unno
- Department of Surgery, National Hospital Organization Sendai Medical Center, Sendai, Miyagi, 983-8520, Japan.,Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8574, Japan
| |
Collapse
|
37
|
Sklirou AD, Gianniou DD, Karousi P, Cheimonidi C, Papachristopoulou G, Kontos CK, Scorilas A, Trougakos IP. High mRNA Expression Levels of Heat Shock Protein Family B Member 2 (HSPB2) Are Associated with Breast Cancer Patients’ Relapse and Poor Survival. Int J Mol Sci 2022; 23:ijms23179758. [PMID: 36077156 PMCID: PMC9456243 DOI: 10.3390/ijms23179758] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
Small heat shock proteins (sHSPs) are ubiquitous ATP-independent chaperones that contribute to the maintenance of proteome integrity and functionality. Recent evidence suggests that sHSPs are ubiquitously expressed in numerous types of tumors and have been proposed to be implicated in oncogenesis and malignant progression. Heat shock protein family B member 2 (HSPB2) is a member of the sHSPs, which is found to be expressed, among others, in human breast cancer cell lines and constitutes an inhibitor of apical caspase activation in the extrinsic apoptotic pathway. In this study, we investigated the potential prognostic significance of HSPB2 mRNA expression levels in breast cancer, which represents the most frequent malignancy in females and one of the three most common cancer types worldwide. To this end, malignant breast tumors along with paired non-cancerous breast tissue specimens were used. HSPB2 expression levels were quantified in these two cohorts using a sensitive and accurate SYBR green-based quantitative real-time polymerase chain reaction (q-RT-PCR). Extensive biostatistical analyses were performed including Kaplan–Meier and Cox regression survival analyses for the assessment of the results. The significant downregulation of HSPB2 gene expression was revealed in breast tumors compared to their adjacent non-cancerous breast tissues. Notably, high HSPB2 mRNA expression predicts poor disease-free survival and overall survival of breast cancer patients. Multivariate Cox regression analysis revealed that HSPB2 mRNA overexpression is a significant predictor of poor prognosis in breast cancer, independent of other clinicopathological factors. In conclusion, high HSPB2 mRNA expression levels are associated with breast cancer patients’ relapse and poor survival.
Collapse
Affiliation(s)
- Aimilia D. Sklirou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Despoina D. Gianniou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Paraskevi Karousi
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece
| | - Christina Cheimonidi
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | | | - Christos K. Kontos
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece
- Correspondence: (A.S.); (I.P.T.); Tel.: +30-210-727-4306 (A.S.); +30-210-727-4555 (I.P.T.)
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
- Correspondence: (A.S.); (I.P.T.); Tel.: +30-210-727-4306 (A.S.); +30-210-727-4555 (I.P.T.)
| |
Collapse
|
38
|
Negahdary M, Angnes L. An aptasensing platform for detection of heat shock protein 70 kDa (HSP70) using a modified gold electrode with lady fern-like gold (LFG) nanostructure. Talanta 2022; 246:123511. [DOI: 10.1016/j.talanta.2022.123511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 10/18/2022]
|
39
|
Peinado-Ruiz IC, Burgos-Molina AM, Sendra-Portero F, Ruiz-Gómez MJ. Relationship between heat shock proteins and cellular resistance to drugs and ageing. Exp Gerontol 2022; 167:111896. [PMID: 35870754 DOI: 10.1016/j.exger.2022.111896] [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: 01/17/2022] [Revised: 07/06/2022] [Accepted: 07/12/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND AND AIMS Ageing is a multifactorial degenerative process which causes a decrease in the cellular capacity for repair and adaptation to external stressors. In this way, it is important to maintain the proper balance of the proteome. Heat shock proteins (HSP) will intervene in this balance, which are responsible for the correct assembly, folding and translocation of other proteins when cells are subjected to stressors. This type of protein is overexpressed in human tumor cells, while its deficit, both in function and quantity, contributes to ageing processes. The present work aims to analyze the response of cells from studies carried out in normal and tumor cells that are subjected to stressors. METHODS AND RESULTS A PubMed search was performed using the keywords "cell ageing, cell longevity, resistance, HSP, heat shock proteins, thermal shock proteins". This search generated 212 articles. Subsequently, a series of inclusion and exclusion criteria were applied to select the articles of interest to be evaluated. Normal cells subjected to external stressors at low doses increase the number of HSP, causing them to become more resistant. In addition, tumor cells expressing high levels of HSP show greater resistance to treatment and increased cell replication. HSP intervene in the cellular resistance of both normal and tumor cells. CONCLUSIONS In the case of normal cells, the increase in HSP levels makes them respond effectively to an external stressor, increasing their resistance and not causing cell death. In the case of tumor cells, there is an increase in resistance to treatment.
Collapse
Affiliation(s)
- Isabel C Peinado-Ruiz
- Departamento de Radiología y Medicina Física, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Antonio M Burgos-Molina
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Francisco Sendra-Portero
- Departamento de Radiología y Medicina Física, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Miguel J Ruiz-Gómez
- Departamento de Radiología y Medicina Física, Facultad de Medicina, Universidad de Málaga, Málaga, Spain.
| |
Collapse
|
40
|
DİRİCAN E, ÇINAR İ. Gossypin'in farklı kanser hücre dizilerinde HSP60 ve HSP70'in gen ekspresyonu üzerindeki etkisi. CUKUROVA MEDICAL JOURNAL 2022. [DOI: 10.17826/cumj.1052787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Amaç: Bu çalışmanın amacı, gossypin'in farklı kanser hücre hatlarında ısı şok proteinleri (HSP) genlerinin ekspresyon seviyesi üzerindeki etkisini incelemektir.
Gereç ve Yöntem: Hücreler, standart kültür koşulları altında büyütüldü. Kanser hücreleri, farklı konsantrasyonlarda (5-100 µg/ml) gossypin ve pozitif kontrol olarak sisplatin (50 µM) ile muamele edildi. Gossypin'in hücre canlılığı ve etkili doz aralığı (5-100 µg/ml), 24, 48 ve 72. saatlerde MTT ile belirlendi. RNA izolasyonu ve cDNA sentezinden sonra, HSP60 ve HSP70 gen ekpresyon seviyesi RT-PCR ile analiz edildi. Gen ekspresyonu için 2-∆∆ct methodu kullanıldı.
Bulgular: MTT sonuçlarına göre kanser hücre hatlarında 25-50-100 µg/ml gossipin dozlarının HSP60 ve HSP70 gen ekspresyon seviyeleri üzerinde etkili olduğu bulundu. Gossypin, üç hücre hattında HSP60 ve HSP70'in ekspresyonunu doza bağımlı olarak etkilemiştir. Üç hücre hattında, 50 µg/ml ve 100 µg/ml gossipin dozları, HSP60 ve HSP70'in ekspresyonunu kontrol grubuna kıyasla önemli ölçüde azalttı.
Sonuç: Sonuçlarımız, farklı hücre dizilerinde çeşitli dozlarda gossypinin antikarsinojenik etkisini güçlü bir şekilde desteklemektedir. Fakat, daha fazla in vivo araştırma ve insan çalışmalarına ihtiyaç olduğuna inanıyoruz. Bulgularımız, gossypin'nin farklı kanser türlerinin önlenmesi ve/veya tedavisi için yeni stratejiler geliştirmek için daha ileri araştırmalar için uygun aday ajan olabileceğini düşündürmektedir.
Collapse
Affiliation(s)
- Ebubekir DİRİCAN
- BAYBURT ÜNİVERSİTESİ, BAYBURT SAĞLIK HİZMETLERİ MESLEK YÜKSEKOKULU
| | | |
Collapse
|
41
|
Özgür A, Kaplan Ö, Gökşen Tosun N, Türkekul İ, Gökçe İ. Green synthesis of silver nanoparticles using Macrolepiota procera extract and investigation of their HSP27, HSP70, and HSP90 inhibitory potentials in human cancer cells. PARTICULATE SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1080/02726351.2022.2089303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Aykut Özgür
- Department of Veterinary Medicine, Laboratory and Veterinary Health Program, Artova Vocational School, Tokat Gaziosmanpaşa University, Tokat, Turkey
| | - Özlem Kaplan
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Nazan Gökşen Tosun
- Department of Biomaterials and Tissue Engineering, Institute of Graduate Studies, Tokat Gaziosmanpaşa University, Tokat, Turkey
| | - İbrahim Türkekul
- Department of Biology, Faculty of Arts and Sciences, Tokat Gaziosmanpaşa University, Tokat, Turkey
| | - İsa Gökçe
- Department of Bioengineering, Faculty of Engineering and Architecture, Tokat Gaziosmanpaşa University, Tokat, Turkey
| |
Collapse
|
42
|
Yi K, Zhou Y, Zhang M, Guo Y. The Core Mechanism of Yiqi Yangjing Decoction Inhibiting Nonsmall-Cell Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2256671. [PMID: 35586682 PMCID: PMC9110163 DOI: 10.1155/2022/2256671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/22/2022] [Indexed: 11/23/2022]
Abstract
Background Yiqi Yangjing prescription (YQYJ) is a traditional Chinese medicine prescription used for treating lung cancer. It has a significant effect on enhancing efficacy, reducing toxic symptoms, and improving patients' physical well-being. The effective inhibitory effect on nonsmall-cell lung cancer (NSCLC) has been demonstrated in vitro and in vivo. However, the mechanism of action and the material basis still remain unclear. Methods In this study, we explored this mechanism using network pharmacology, after which we explored the pharmacodynamics and the action mechanism of YQYJ using cell viability evaluation, plate clone formation assay, flow cytometry, real-time quantitative PCR, and Western blot. Results The enrichment results showed that there were 50 active components and 68 core targets related to YQYJ inhibiting NSCLC, including quercetin, luteolin, gamatin, kaempferol, heat shock protein HSP 90-alpha (HSP90AA1), cyclin-dependent kinase 2 (CDK2), epidermal growth factor receptor (EGFR), signal transducer and activator of transcription 3 (STAT3), and others. Among them, quercetin and kaempferol revealed the best binding effect with core targets. Most importantly, YQYJ promoted A549 cells from the quiescent phase into the proliferative phase to enhance the sensitivity of A549 cells to YQYJ and inhibited the proliferation of A549 cells significantly (P < 0.05). The A549 cells were blocked in both S and G2/M phases while the apoptosis ratio was increased. The proliferation score of A549 cells treated with YQYJ was significantly reduced compared to A549 cells in the proliferative phase (P < 0.05). This regulatory effect was related to the expression regulation of HSP90AA1, CDK2, STAT3, and phosphor-STAT3 (p-STAT3) by YQYJ, kaempferol, and quercetin. Conclusion Our results suggested that the inhibition of NSCLC via YQYJ had multicomponent and multitarget characteristics. Its core mechanism is related to the regulation of the cell cycle, proliferation, and apoptosis of NSCLC. This study provides a direction and scientific basis for exploring the future mechanism of YQYJ for the treatment of NSCLC.
Collapse
Affiliation(s)
- Kaiyan Yi
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yaning Zhou
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ming Zhang
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yijun Guo
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
43
|
Mattos DR, Weinman MA, Wan X, Goodall CP, Serrill JD, McPhail KL, Milovancev M, Bracha S, Ishmael JE. Canine osteosarcoma cells exhibit basal accumulation of multiple chaperone proteins and are sensitive to small molecule inhibitors of GRP78 and heat shock protein function. Cell Stress Chaperones 2022; 27:223-239. [PMID: 35244890 PMCID: PMC9106791 DOI: 10.1007/s12192-022-01263-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma is the most common type of bone cancer in dogs and humans, with significant numbers of patients experiencing treatment failure and disease progression. In our search for new approaches to treat osteosarcoma, we previously detected multiple chaperone proteins in the surface-exposed proteome of canine osteosarcoma cells. In the present study, we characterized expression of representative chaperones and find evidence for stress adaptation in canine osteosarcoma cells relative to osteogenic progenitors from normal bone. We compared the cytotoxic potential of direct (HA15) and putative (OSU-03012) inhibitors of Grp78 function and found canine POS and HMPOS osteosarcoma cells to be more sensitive to both compounds than normal cells. HA15 and OSU-03012 increased the thermal stability of Grp78 in intact POS cells at low micromolar concentrations, but each induced distinct patterns in Grp78 expression without significant change in Grp94. Both inhibitors were as effective alone as carboplatin and showed little evidence of synergy in combination treatment. However, HMPOS cells with acquired resistance to carboplatin were sensitive to inhibition of Grp78 (by HA15; OSU-03012), Hsp70 (by VER-155008), and Hsp90 (by 17-AAG) function. These results suggest that multiple nodes within the osteosarcoma chaperome may be relevant chemotherapeutic targets against platinum resistance.
Collapse
Affiliation(s)
- Daphne R Mattos
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 411 Pharmacy Building, Corvallis, OR, 97331, USA
| | - Marcus A Weinman
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, USA
- CMB Graduate Program, Department of Neurological Sciences, University of Vermont, Burlington, VT, 05405, USA
| | - Xuemei Wan
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 411 Pharmacy Building, Corvallis, OR, 97331, USA
| | - Cheri P Goodall
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, USA
| | - Jeffrey D Serrill
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 411 Pharmacy Building, Corvallis, OR, 97331, USA
| | - Kerry L McPhail
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 411 Pharmacy Building, Corvallis, OR, 97331, USA
| | - Milan Milovancev
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, USA.
| | - Shay Bracha
- College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA.
| | - Jane E Ishmael
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 411 Pharmacy Building, Corvallis, OR, 97331, USA.
| |
Collapse
|
44
|
Yu Y, Wang B, Sun M, Zhang Y, Hou L, Wang S, Chen T, Yang F, Ma Z. Lysosomal activable Vorinostat carrier-prodrug self-assembling with BPQDs enables photothermal oncotherapy to reverse tumor thermotolerance and metastasis. Int J Pharm 2022; 617:121580. [PMID: 35202725 DOI: 10.1016/j.ijpharm.2022.121580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/27/2022] [Accepted: 02/10/2022] [Indexed: 12/16/2022]
Abstract
Photothermal therapy (PTT) is becoming increasing prevalent in clinic for eradicating the primary tumor and improving cancer patients' compliance. However, photothermal resistance and distal metastasis still haunt the tumor treatment with PTT. Herein, on the basis that histone deacetylase acetylase inhibitor (HDACis) could activate the expression of anti-tumor gene and accelerate the differentiation and apoptosis of tumor cells, we propose that HDACis supplementing PTT could overcome those obstacles with appropriate drug-controlled release strategy. Thus, we fabricated a nano-complex of lysosomal activable vorinostat (SAHA) carrier-prodrug encapsulating black phosphorus quantum dots (BPQDs@PPS) to counter those challenges in PTT. With spherical morphology and favorable bio-safety, BPQDs@PPS could release BPQDs and Vorinostat spontaneously in lysosome, not only effectively inhibiting tumor growth, but also reversing tumor thermotolerance and metastasis within a PTT procedure. Especially, both western blot and immunofluorescence analysis validate that Vorinostat enables PTT to reverse tumor thermotolerance and distal metastasis by down-regulation of HSP70 and up-regulation of H3. Therefore, this research not only reveals the mechanism how HDACis supplement PTT in reversing tumor thermotolerance and metastasis, but also provides a promising prospect to upgrade clinical photothermal therapy.
Collapse
Affiliation(s)
- Yingjie Yu
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Bingkai Wang
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Miao Sun
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Yunchang Zhang
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Lei Hou
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Sizhen Wang
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Tianheng Chen
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Feng Yang
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Zhiqiang Ma
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China.
| |
Collapse
|
45
|
Chee WKD, Yeoh JW, Dao VL, Poh CL. Thermogenetics: Applications come of age. Biotechnol Adv 2022; 55:107907. [PMID: 35041863 DOI: 10.1016/j.biotechadv.2022.107907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/13/2021] [Accepted: 01/09/2022] [Indexed: 12/20/2022]
Abstract
Temperature is a ubiquitous physical cue that is non-invasive, penetrative and easy to apply. In the growing field of thermogenetics, through beneficial repurposing of natural thermosensing mechanisms, synthetic biology is bringing new opportunities to design and build robust temperature-sensitive (TS) sensors which forms a thermogenetic toolbox of well characterised biological parts. Recent advancements in technological platforms available have expedited the discovery of novel or de novo thermosensors which are increasingly deployed in many practical temperature-dependent biomedical, industrial and biosafety applications. In all, the review aims to convey both the exhilarating recent technological developments underlying the advancement of thermosensors and the exciting opportunities the nascent thermogenetic field holds for biomedical and biotechnology applications.
Collapse
Affiliation(s)
- Wai Kit David Chee
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
| | - Jing Wui Yeoh
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
| | - Viet Linh Dao
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
| | - Chueh Loo Poh
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore.
| |
Collapse
|
46
|
Unravelling the Interaction of Piperlongumine with the Nucleotide-Binding Domain of HSP70: A Spectroscopic and In Silico Study. Pharmaceuticals (Basel) 2021; 14:ph14121298. [PMID: 34959698 PMCID: PMC8703466 DOI: 10.3390/ph14121298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022] Open
Abstract
Piperlongumine (PPL) is an alkaloid extracted from several pepper species that exhibits anti-inflammatory and anti-carcinogenic properties. Nevertheless, the molecular mode of action of PPL that confers such powerful pharmacological properties remains unknown. From this perspective, spectroscopic methods aided by computational modeling were employed to characterize the interaction between PPL and nucleotide-binding domain of heat shock protein 70 (NBD/HSP70), which is involved in the pathogenesis of several diseases. Steady-state fluorescence spectroscopy along with time-resolved fluorescence revealed the complex formation based on a static quenching mechanism. Van't Hoff analyses showed that the binding of PPL toward NBD is driven by equivalent contributions of entropic and enthalpic factors. Furthermore, IDF and Scatchard methods applied to fluorescence intensities determined two cooperative binding sites with Kb of (6.3 ± 0.2) × 104 M-1. Circular dichroism determined the thermal stability of the NBD domain and showed that PPL caused minor changes in the protein secondary structure. Computational simulations elucidated the microenvironment of these interactions, showing that the binding sites are composed mainly of polar amino acids and the predominant interaction of PPL with NBD is Van der Waals in nature.
Collapse
|
47
|
Chen C, Wang YS, Zhang ET, Li GA, Liu WY, Li Y, Jin YH. (20S) Ginsenoside Rh2 Exerts Its Anti-Tumor Effect by Disrupting the HSP90A-Cdc37 System in Human Liver Cancer Cells. Int J Mol Sci 2021; 22:ijms222313170. [PMID: 34884975 PMCID: PMC8658384 DOI: 10.3390/ijms222313170] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/22/2022] Open
Abstract
(20S) ginsenoside Rh2 (G-Rh2), a major bioactive metabolite of ginseng, effectively inhibits the survival and proliferation of human liver cancer cells. However, its molecular targets and working mechanism remain largely unknown. Excitingly, we screened out heat shock protein 90 alpha (HSP90A), a key regulatory protein associated with liver cancer, as a potential target of (20S) G-Rh2 by phage display analysis and mass spectrometry. The molecular docking and thermal shift analyses demonstrated that (20S) G-Rh2 directly bound to HSP90A, and this binding was confirmed to inhibit the interaction between HSP90A and its co-chaperone, cell division cycle control protein 37 (Cdc37). It is well-known that the HSP90A-Cdc37 system aids in the folding and maturation of cyclin-dependent kinases (CDKs). As expected, CDK4 and CDK6, the two G0-G1 phase promoting kinases as well as CDK2, a key G1-S phase transition promoting kinase, were significantly downregulated with (20S) G-Rh2 treatment, and these downregulations were mediated by the proteasome pathway. In the same condition, the cell cycle was arrested at the G0-G1 phase and cell growth was inhibited significantly by (20S) G-Rh2 treatment. Taken together, this study for the first time reveals that (20S) G-Rh2 exerts its anti-tumor effect by targeting HSP90A and consequently disturbing the HSP90A-Cdc37 chaperone system. HSP90A is frequently overexpressed in human hepatoma cells and the higher expression is closely correlated to the poor prognosis of liver cancer patients. Thus, (20S) G-Rh2 might become a promising alternative drug for liver cancer therapy.
Collapse
|
48
|
The Metastasis Suppressor NDRG1 Directly Regulates Androgen Receptor Signaling in Prostate Cancer. J Biol Chem 2021; 297:101414. [PMID: 34785213 PMCID: PMC8668986 DOI: 10.1016/j.jbc.2021.101414] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
N-myc-downregulated gene 1 (NDRG1) has potent anticancer effects and inhibits cell growth, survival, metastasis, and angiogenesis. Previous studies suggested that NDRG1 is linked to the androgen signaling network, but this mechanistic relationship is unclear. Considering the crucial role of the androgen receptor (AR) in prostate cancer (PCa) progression, here we examined for the first time the effect of NDRG1 on AR expression, activation, and downstream signaling in LNCaP, 22Rv1, and C4-2B PCa cell types. We demonstrate that NDRG1 effectively promotes interaction of AR with the chaperone HSP90, which in turn stabilizes the AR while decreasing its androgen-mediated activation. The expression of NDRG1 suppressed: (1) AR activation, as measured by p-ARSer213 and p-ARSer81; (2) expression of a major AR transcriptional target, prostate-specific antigen (PSA); and (3) AR transcriptional activity, probably via inhibiting the c-Jun-AR interaction by reducing c-Jun phosphorylation (p-c-JunSer63). NDRG1 was also demonstrated to inhibit multiple key molecules involved in androgen-dependent and -independent signaling (namely EGFR, HER2, HER3, PI3K, STAT3, and NF-κB), which promote the development of castration-resistant prostate cancer. We also identified the cysteine-rich secretory protein/antigen 5/pathogenesis related-1 (CAP) domain of NDRG1 as vital for inhibition of AR activity. Examining NDRG1 and p-NDRG1 in PCa patient specimens revealed a significant negative correlation between NDRG1 and PSA levels in prostatectomy patients that went on to develop metastasis. These results highlight a vital role for NDRG1 in androgen signaling and its potential as a key therapeutic target and biomarker in PCa.
Collapse
|
49
|
With or without You: Co-Chaperones Mediate Health and Disease by Modifying Chaperone Function and Protein Triage. Cells 2021; 10:cells10113121. [PMID: 34831344 PMCID: PMC8619055 DOI: 10.3390/cells10113121] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 01/18/2023] Open
Abstract
Heat shock proteins (HSPs) are a family of molecular chaperones that regulate essential protein refolding and triage decisions to maintain protein homeostasis. Numerous co-chaperone proteins directly interact and modify the function of HSPs, and these interactions impact the outcome of protein triage, impacting everything from structural proteins to cell signaling mediators. The chaperone/co-chaperone machinery protects against various stressors to ensure cellular function in the face of stress. However, coding mutations, expression changes, and post-translational modifications of the chaperone/co-chaperone machinery can alter the cellular stress response. Importantly, these dysfunctions appear to contribute to numerous human diseases. Therapeutic targeting of chaperones is an attractive but challenging approach due to the vast functions of HSPs, likely contributing to the off-target effects of these therapies. Current efforts focus on targeting co-chaperones to develop precise treatments for numerous diseases caused by defects in protein quality control. This review focuses on the recent developments regarding selected HSP70/HSP90 co-chaperones, with a concentration on cardioprotection, neuroprotection, cancer, and autoimmune diseases. We also discuss therapeutic approaches that highlight both the utility and challenges of targeting co-chaperones.
Collapse
|