1
|
Mamun M, Zheng YC, Wang N, Wang B, Zhang Y, Pang JR, Shen DD, Liu HM, Gao Y. Decoding CLU (Clusterin): Conquering cancer treatment resistance and immunological barriers. Int Immunopharmacol 2024; 137:112355. [PMID: 38851158 DOI: 10.1016/j.intimp.2024.112355] [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: 04/25/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/10/2024]
Abstract
One major obstacle in the treatment of cancer is the presence of proteins resistant to cancer therapy, which can impede the effectiveness of traditional approaches such as radiation and chemotherapy. This resistance can lead to disease progression and cause treatment failure. Extensive research is currently focused on studying these proteins to create tailored treatments that can circumvent resistance mechanisms. CLU (Clusterin), a chaperone protein, has gained notoriety for its role in promoting resistance to a wide range of cancer treatments, including chemotherapy, radiation therapy, and targeted therapy. The protein has also been discovered to have a role in regulating the immunosuppressive environment within tumors. Its ability to influence oncogenic signaling and inhibit cell death bolster cancer cells resistant against treatments, which poses a significant challenge in the field of oncology. Researchers are actively investigating to the mechanisms by which CLU exerts its resistance-promoting effects, with the ultimate goal of developing strategies to circumvent its impact and enhance the effectiveness of cancer therapies. By exploring CLU's impact on cancer, resistance mechanisms, tumor microenvironment (TME), and therapeutic strategies, this review aims to contribute to the ongoing efforts to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Maa Mamun
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ning Wang
- The School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Bo Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yu Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Jing-Ru Pang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Dan-Dan Shen
- Key Laboratory of Endometrial Disease Prevention and Treatment, Zhengzhou China, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
2
|
Yue W, Li X, Zhan X, Wang L, Ma J, Bi M, Wang Q, Gu X, Xie B, Liu T, Guo H, Zhu X, Song C, Qiao J, Li M. PARP inhibitors suppress tumours via centrosome error-induced senescence independent of DNA damage response. EBioMedicine 2024; 103:105129. [PMID: 38640836 PMCID: PMC11052917 DOI: 10.1016/j.ebiom.2024.105129] [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: 08/10/2023] [Revised: 04/07/2024] [Accepted: 04/07/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Poly(ADP-ribose) polymerase (PARP) inhibitors have emerged as promising chemotherapeutic drugs primarily against BRCA1/2-associated tumours, known as synthetic lethality. However, recent clinical trials reported patients' survival benefits from PARP inhibitor treatments, irrelevant to homologous recombination deficiency. Therefore, revealing the therapeutic mechanism of PARP inhibitors beyond DNA damage repair is urgently needed, which can facilitate precision medicine. METHODS A CRISPR-based knock-in technology was used to establish stable BRCA1 mutant cancer cells. The effects of PARP inhibitors on BRCA1 mutant cancer cells were evaluated by biochemical and cell biological experiments. Finally, we validated its in vivo effects in xenograft and patient-derived xenograft (PDX) tumour mice. FINDINGS In this study, we uncovered that the majority of clinical BRCA1 mutations in breast cancers were in and near the middle of the gene, rather than in essential regions for DNA damage repair. Representative mutations such as R1085I and E1222Q caused transient extra spindle poles during mitosis in cancer cells. PAR, which is synthesized by PARP2 but not PARP1 at mitotic centrosomes, clustered these transient extra poles, independent of DNA damage response. Common PARP inhibitors could effectively suppress PARP2-synthesized PAR and induce cell senescence by abrogating the correction of mitotic extra-pole error. INTERPRETATION Our findings uncover an alternative mechanism by which PARP inhibitors efficiently suppress tumours, thereby pointing to a potential new therapeutic strategy for centrosome error-related tumours. FUNDING Funded by National Natural Science Foundation of China (NSFC) (T2225006, 82272948, 82103106), Beijing Municipal Natural Science Foundation (Key program Z220011), and the National Clinical Key Specialty Construction Program, P. R. China (2023).
Collapse
Affiliation(s)
- Wei Yue
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology (Peking University Third Hospital), Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Xinyu Li
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology (Peking University Third Hospital), Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Xiaolu Zhan
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Lei Wang
- Centre for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Peking-Tsinghua Centre for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jihong Ma
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology (Peking University Third Hospital), Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Meiyu Bi
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology (Peking University Third Hospital), Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Qilong Wang
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology (Peking University Third Hospital), Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Xiaoyang Gu
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology (Peking University Third Hospital), Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Bingteng Xie
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Tong Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Hongyan Guo
- National Clinical Research Centre for Obstetrics and Gynaecology (Peking University Third Hospital), Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Xin Zhu
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology (Peking University Third Hospital), Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Chen Song
- Centre for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Peking-Tsinghua Centre for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology (Peking University Third Hospital), Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Mo Li
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology (Peking University Third Hospital), Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| |
Collapse
|
3
|
Nikesitch N, Beraldi E, Zhang F, Adomat H, Bell R, Suzuki K, Fazli L, Hy Kung S, Wells C, Pinette N, Saxena N, Wang Y, Gleave M. Chaperone-mediated autophagy promotes PCa survival during ARPI through selective proteome remodeling. Oncogene 2023; 42:748-758. [PMID: 36611121 DOI: 10.1038/s41388-022-02573-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 01/09/2023]
Abstract
The androgen receptor (AR) plays an important role in PCa metabolism, with androgen receptor pathway inhibition (ARPI) subjecting PCa cells to acute metabolic stress caused by reduced biosynthesis and energy production. Defining acute stress response mechanisms that alleviate ARPI stress and therefore mediate prostate cancer (PCa) treatment resistance will help improve therapeutic outcomes of patients treated with ARPI. We identified the up-regulation of chaperone-mediated autophagy (CMA) in response to acute ARPI stress, which persisted in castration-resistant PCa (CRPC); previously undefined in PCa. CMA is a selective protein degradation pathway and a key stress response mechanism up-regulated under several stress stimuli, including metabolic stress. Through selective protein degradation, CMA orchestrates the cellular stress response by regulating cellular pathways through selective proteome remodeling. Through broad-spectrum proteomic analysis, CMA coordinates metabolic reprogramming of PCa cells to sustain PCa growth and survival during ARPI; through the upregulation of mTORC1 signaling and pathways associated with PCa biosynthesis and energetics. This not only promoted PCa growth during ARPI, but also promoted the emergence of CRPC in-vivo. During CMA inhibition, PCa metabolism is compromised, leading to ATP depletion, resulting in a profound anti-proliferative effect on PCa cells, and is enhanced when combined with ARPI. Furthermore, CMA inhibition prevented in-vivo tumour formation, and also re-sensitized enzalutamide-resistant cell lines in-vitro. The profound anti-proliferative effect of CMA inhibition was attributed to cell cycle arrest mediated through p53 transcriptional repression of E2F target genes. In summary, CMA is an acute ARPI stress response mechanism, essential in alleviating ARPI induced metabolic stress, essential for ensuring PCa growth and survival. CMA plays a critical role in the development of ARPI resistance in PCa.
Collapse
Affiliation(s)
- Nicholas Nikesitch
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Eliana Beraldi
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Fan Zhang
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Hans Adomat
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Robert Bell
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Kotaro Suzuki
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Sonia Hy Kung
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Christopher Wells
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Nicholas Pinette
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Neetu Saxena
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada.
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
4
|
Reformation of the chondroitin sulfate glycocalyx enables progression of AR-independent prostate cancer. Nat Commun 2022; 13:4760. [PMID: 35963852 PMCID: PMC9376089 DOI: 10.1038/s41467-022-32530-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/03/2022] [Indexed: 11/09/2022] Open
Abstract
Lineage plasticity of prostate cancer is associated with resistance to androgen receptor (AR) pathway inhibition (ARPI) and supported by a reactive tumor microenvironment. Here we show that changes in chondroitin sulfate (CS), a major glycosaminoglycan component of the tumor cell glycocalyx and extracellular matrix, is AR-regulated and promotes the adaptive progression of castration-resistant prostate cancer (CRPC) after ARPI. AR directly represses transcription of the 4-O-sulfotransferase gene CHST11 under basal androgen conditions, maintaining steady-state CS in prostate adenocarcinomas. When AR signaling is inhibited by ARPI or lost during progression to non-AR-driven CRPC as a consequence of lineage plasticity, CHST11 expression is unleashed, leading to elevated 4-O-sulfated chondroitin levels. Inhibition of the tumor cell CS glycocalyx delays CRPC progression, and impairs growth and motility of prostate cancer after ARPI. Thus, a reactive CS glycocalyx supports adaptive survival and treatment resistance after ARPI, representing a therapeutic opportunity in patients with advanced prostate cancer. Chondroitin sulfate (CS) is one of the most abundant glycosaminoglycans in prostate cancers. Here the authors show that inhibition of the androgen receptor pathway leads to the upregulation of CS, which promotes prostate cancer growth and metastasis.
Collapse
|
5
|
Yue W, Ma J, Xiao Y, Wang P, Gu X, Xie B, Li M. The Apoptotic Resistance of BRCA1-Deficient Ovarian Cancer Cells is Mediated by cAMP. Front Cell Dev Biol 2022; 10:889656. [PMID: 35517499 PMCID: PMC9065249 DOI: 10.3389/fcell.2022.889656] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/23/2022] [Indexed: 11/17/2022] Open
Abstract
Breast cancer type 1 susceptibility protein (BRCA1) is essential for homologous recombination repair of DNA double-strand breaks. Loss of BRCA1 is lethal to embryos due to extreme genomic instability and the activation of p53-dependent apoptosis. However, the apoptosis is resisted in BRCA1-deficient cancer cells even though their p53 is proficient. In this study, by analysis of transcriptome data of ovarian cancer patients bearing BRCA1 defects in TCGA database, we found that cAMP signaling pathway was significantly activated. Experimentally, we found that BRCA1 deficiency caused an increased expression of ADRB1, a transmembrane receptor that can promote the generation of cAMP. The elevated cAMP not only inhibited DNA damage-induced apoptosis through abrogating p53 accumulation, but also suppressed the proliferation of cytotoxic T lymphocytes by enhancing the expression of immunosuppressive factors DKK1. Inhibition of ADRB1 effectively killed cancer cells by abolishing the apoptotic resistance. These findings uncover a novel mechanism of apoptotic resistance in BRCA1-deficient ovarian cancer cells and point to a potentially new strategy for treating BRCA1-mutated tumors.
Collapse
Affiliation(s)
- Wei Yue
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, China
| | - Jihong Ma
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, China
| | - Yinan Xiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, China
| | - Pan Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, China
| | - Xiaoyang Gu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, China
| | - Bingteng Xie
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Mo Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, China
- *Correspondence: Mo Li,
| |
Collapse
|
6
|
Andújar-Vera F, García-Fontana C, Sanabria-de la Torre R, González-Salvatierra S, Martínez-Heredia L, Iglesias-Baena I, Muñoz-Torres M, García-Fontana B. Identification of Potential Targets Linked to the Cardiovascular/Alzheimer's Axis through Bioinformatics Approaches. Biomedicines 2022; 10:389. [PMID: 35203598 PMCID: PMC8962298 DOI: 10.3390/biomedicines10020389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 12/23/2022] Open
Abstract
The identification of common targets in Alzheimer's disease (AD) and cardiovascular disease (CVD) in recent years makes the study of the CVD/AD axis a research topic of great interest. Besides aging, other links between CVD and AD have been described, suggesting the existence of common molecular mechanisms. Our study aimed to identify common targets in the CVD/AD axis. For this purpose, genomic data from calcified and healthy femoral artery samples were used to identify differentially expressed genes (DEGs), which were used to generate a protein-protein interaction network, where a module related to AD was identified. This module was enriched with the functionally closest proteins and analyzed using different centrality algorithms to determine the main targets in the CVD/AD axis. Validation was performed by proteomic and data mining analyses. The proteins identified with an important role in both pathologies were apolipoprotein E and haptoglobin as DEGs, with a fold change about +2 and -2, in calcified femoral artery vs healthy artery, respectively, and clusterin and alpha-2-macroglobulin as close interactors that matched in our proteomic analysis. However, further studies are needed to elucidate the specific role of these proteins, and to evaluate its function as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Francisco Andújar-Vera
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Department of Computer Science and Artificial Intelligence, University of Granada, 18071 Granada, Spain
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI Institute), 18014 Granada, Spain
| | - Cristina García-Fontana
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- CIBERFES, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Raquel Sanabria-de la Torre
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Sheila González-Salvatierra
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Luis Martínez-Heredia
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Iván Iglesias-Baena
- Fundación para la Investigación Biosanitaria de Andalucía Oriental-Alejandro Otero (FIBAO), 18012 Granada, Spain;
| | - Manuel Muñoz-Torres
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- CIBERFES, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Beatriz García-Fontana
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- CIBERFES, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
7
|
|
8
|
Sreedurgalakshmi K, Srikar R, Harikrishnan K, Srinivasan L, Rajkumari R. Cetuximab-siRNA Conjugate Linked Through Cationized Gelatin Knocks Down KRAS G12C Mutation in NSCLC Sensitizing the Cells Toward Gefitinib. Technol Cancer Res Treat 2021; 20:15330338211041453. [PMID: 34542333 PMCID: PMC8461128 DOI: 10.1177/15330338211041453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Delivery of small-interfering RNA (siRNA) has been of great interest in the past decade for effective gene silencing. To overcome synthetic and regulatory challenges posed by nanoparticle-mediated siRNA delivery, antibody–siRNA conjugate (ARC) platform is emerging as a potential siRNA delivery system suitable for clinical translation. Herein, we have developed a delivery technology based on the ARC platform for stable delivery of siRNA called as Gelatin-Antibody Delivery System (GADS). In GADS, positively charged gelatin acts as a linker between antibody–siRNA and enables the endosomal escape of siRNA for gene silencing postcellular internalization. For proof of concept, we synthesized a scalable GADS conjugate comprising of Cetuximab (CTB), cationized gelatin (cGel) and NSCLC KRASG12C-specific siRNA. CTB was chemically conjugated to cGel through an amide link to form the CTB–cGel complex. Thereafter, siRNA was chemically conjugated to the cGel moiety of the complex through the thioether link to form CTB–cGel–siRNA conjugate. RP-HPLC analysis was used to monitor the reaction while gel retardation assay was used to determine siRNA loading capacity. SPR analysis showed the preservation of ligand binding affinity of antibody conjugates with KD of ∼0.3 nM. Furthermore, cellular internalization study using florescent microscopy revealed receptor-mediated endocytosis. The conjugate targeted EGFR receptor of KRAS mutant NSCLC to specifically knockdown G12C mutation. The oncogene knockdown sensitized the cells toward small molecule inhibitor—Gefitinib causing ∼70% loss in cell viability. Western blot analysis revealed significant downregulation for various RAS downstream proteins postoncogene knockdown. Comparison of the efficiency of GADS vis-à-vis positive siRNA control and CRISPR–Cas9-based knockout of KRAS Exon 2 in the NCI-H23 NSCLC cell line suggests GADS as a potential technology for clinical translation of gene therapy.
Collapse
Affiliation(s)
- K. Sreedurgalakshmi
- Vellore Institute of Technology, Vellore, Tamil Nadu, India
- R&D, Levim Biotech LLP, Chennai, Tamil Nadu, India
| | - R. Srikar
- R&D, Levim Biotech LLP, Chennai, Tamil Nadu, India
- R. Srikar, Division of Biosimilars and Gene Therapy, R&D,
Levim Biotech LLP, Chennai, Tamil Nadu, India.
Reena Rajkumari, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India.
| | | | | | | |
Collapse
|
9
|
Pandey M, Cuddihy G, Gordon JA, Cox ME, Wasan KM. Inhibition of Scavenger Receptor Class B Type 1 (SR-B1) Expression and Activity as a Potential Novel Target to Disrupt Cholesterol Availability in Castration-Resistant Prostate Cancer. Pharmaceutics 2021; 13:1509. [PMID: 34575583 PMCID: PMC8467449 DOI: 10.3390/pharmaceutics13091509] [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: 06/25/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023] Open
Abstract
There have been several studies that have linked elevated scavenger receptor class b type 1 (SR-B1) expression and activity to the development and progression of castration-resistant prostate cancer (CRPC). SR-B1 facilitates the influx of cholesterol to the cell from lipoproteins in systemic circulation. This influx of cholesterol may be important for many cellular functions, including the synthesis of androgens. Castration-resistant prostate cancer tumors can synthesize androgens de novo to supplement the loss of exogenous sources often induced by androgen deprivation therapy. Silencing of SR-B1 may impact the ability of prostate cancer cells, particularly those of the castration-resistant state, to maintain the intracellular supply of androgens by removing a supply of cholesterol. SR-B1 expression is elevated in CRPC models and has been linked to poor survival of patients. The overarching belief has been that cholesterol modulation, through either synthesis or uptake inhibition, will impact essential signaling processes, impeding the proliferation of prostate cancer. The reduction in cellular cholesterol availability can impede prostate cancer proliferation through both decreased steroid synthesis and steroid-independent mechanisms, providing a potential therapeutic target for the treatment of prostate cancer. In this article, we discuss and highlight the work on SR-B1 as a potential novel drug target for CRPC management.
Collapse
Affiliation(s)
- Mitali Pandey
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| | - Grace Cuddihy
- College of Pharmacy and Nutrition, University of Saskatchewan, 104 Clinic Place, Saskatoon, SK S7N 2Z4, Canada;
| | - Jacob A. Gordon
- Oncology Bioscience, Oncology R&D, AstraZeneca, Boston, MA 02451, USA;
| | - Michael E. Cox
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| | - Kishor M. Wasan
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| |
Collapse
|
10
|
Xu L, Tian S, Peng X, Hua Y, Yang W, Chen L, Liu S, Wu W, Zhao J, He J, Wu L, Yang J, Zheng Y. Clusterin inhibits Aβ 42 aggregation through a "strawberry model" as detected by FRET-FCS. J Neurochem 2021; 158:444-454. [PMID: 33694231 DOI: 10.1111/jnc.15344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/19/2021] [Accepted: 03/07/2021] [Indexed: 11/28/2022]
Abstract
Extracellular plaque deposits of β-amyloid peptide (Aβ) are one of the main pathological features of Alzheimer's disease (AD). The aggregation of Aβ42 species, especially Aβ42 oligomers, is still an active research field in AD pathogenesis. Secretory clusterin protein (sCLU), an extracellular chaperone, plays an important role in AD pathogenesis. Although sCLU interacts directly with Aβ42 in vitro and in vivo, the mechanism is not clear. In this paper, His-tagged sCLU (sCLU-His) was cloned, expressed and purified, and we applied florescence resonance energy transfer-fluorescence correlation spectroscopy (FRET-FCS) to investigate the direct interaction of sCLU-His and Aβ42 at the single-molecule fluorescence level in vitro. Here, we chose four different fluorescently labeled Aβ42 oligomers to form two different groups of aggregation models, easy or difficult to aggregate. The results showed that sCLU-His could form complexes with both aggregation models, and sCLU-His inhibited the aggregation of Aβ42/RB ~ Aβ42/Atto647 (easy to aggregate model). The complexes were produced as the Aβ42/Label adhered to the sCLU-His, which is similar to a "strawberry model," as strawberry seeds are dotted on the outer surface of strawberries. This work provided additional insight into the interaction mechanism of sCLU and Aβ42 .
Collapse
Affiliation(s)
- Lingwan Xu
- School of Sciences, Beijing Jiaotong University, Beijing, China
| | - Shijun Tian
- Hebei Agriculture University, Baoding, China
| | - Xianglei Peng
- School of Sciences, Beijing Jiaotong University, Beijing, China
| | - Ying Hua
- School of Sciences, Beijing Jiaotong University, Beijing, China
| | - Wenxuan Yang
- School of Sciences, Beijing Jiaotong University, Beijing, China
| | - Longwei Chen
- School of Sciences, Beijing Jiaotong University, Beijing, China
| | - Shilei Liu
- School of Sciences, Beijing Jiaotong University, Beijing, China
| | - Wenzheng Wu
- School of Sciences, Beijing Jiaotong University, Beijing, China
| | - Jiang Zhao
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Jinsheng He
- School of Sciences, Beijing Jiaotong University, Beijing, China
| | - Liqing Wu
- National Institute of Metrology, Beijing, China
| | - Jingfa Yang
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Yanpeng Zheng
- School of Sciences, Beijing Jiaotong University, Beijing, China
| |
Collapse
|
11
|
Cheimonidi C, Grivas IN, Sesti F, Kavrochorianou N, Gianniou DD, Taoufik E, Badounas F, Papassideri I, Rizzi F, Tsitsilonis OE, Haralambous S, Trougakos IP. Clusterin overexpression in mice exacerbates diabetic phenotypes but suppresses tumor progression in a mouse melanoma model. Aging (Albany NY) 2021; 13:6485-6505. [PMID: 33744871 PMCID: PMC7993736 DOI: 10.18632/aging.202788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 01/13/2021] [Indexed: 04/24/2023]
Abstract
Clusterin (CLU) is an ATP-independent small heat shock protein-like chaperone, which functions both intra- and extra-cellularly. Consequently, it has been functionally involved in several physiological (including aging), as well as in pathological conditions and most age-related diseases, e.g., cancer, neurodegeneration, and metabolic syndrome. To address CLU function at an in vivo model we established CLU transgenic (Tg) mice bearing ubiquitous or pancreas-targeted CLU overexpression (OE). Our downstream analyses in established Tg lines showed that ubiquitous or pancreas-targeted CLU OE in mice affected antioxidant, proteostatic and metabolic pathways. Targeted OE of CLU in the pancreas, which also resulted in CLU upregulation in the liver likely via systemic effects, increased basal glucose levels in the circulation and exacerbated diabetic phenotypes. Furthermore, by establishing a syngeneic melanoma mouse tumor model we found that ubiquitous CLU OE suppressed melanoma cells growth, indicating a likely tumor suppressor function in early phases of tumorigenesis. Our observations provide in vivo evidence corroborating the notion that CLU is a potential modulator of metabolic and/or proteostatic pathways playing an important role in diabetes and tumorigenesis.
Collapse
Affiliation(s)
- Christina Cheimonidi
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens 15784, Greece
| | - Ioannis N. Grivas
- Inflammation Research Laboratory, Department of Immunology, Transgenic Technology Laboratory, Hellenic Pasteur Institute, Athens 11521, Greece
| | - Fabiola Sesti
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens 15784, Greece
| | - Nadia Kavrochorianou
- Inflammation Research Laboratory, Department of Immunology, Transgenic Technology Laboratory, Hellenic Pasteur Institute, Athens 11521, Greece
| | - Despoina D. Gianniou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens 15784, Greece
| | - Era Taoufik
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens 11521, Greece
| | - Fotis Badounas
- Inflammation Research Laboratory, Department of Immunology, Transgenic Technology Laboratory, Hellenic Pasteur Institute, Athens 11521, Greece
| | - Issidora Papassideri
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens 15784, Greece
| | - Federica Rizzi
- Dipartimento di Medicina e Chirurgia, Universita di Parma, Parma 43125, Italy
- Istituto Nazionale Biostrutture e Biosistemi (I.N.B.B.), Roma 00136, Italy
| | - Ourania E. Tsitsilonis
- Department of Animal and Human Physiology, Faculty of Biology, National and Kapodistrian University of Athens, Athens 15784, Greece
| | - Sylva Haralambous
- Inflammation Research Laboratory, Department of Immunology, Transgenic Technology Laboratory, Hellenic Pasteur Institute, Athens 11521, Greece
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens 15784, Greece
| |
Collapse
|
12
|
Liu Y, Zhou Y, Ma X, Chen L. Inhibition Lysosomal Degradation of Clusterin by Protein Kinase D3 Promotes Triple-Negative Breast Cancer Tumor Growth. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003205. [PMID: 33643800 PMCID: PMC7887572 DOI: 10.1002/advs.202003205] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/30/2020] [Indexed: 06/10/2023]
Abstract
Triple negative breast cancer (TNBC), with its lack of targeted therapies, shows the worst mortality rate among all breast cancer subtypes. Clusterin (CLU) is implicated to play important oncogenic roles in cancer via promoting various downstream oncogenic pathways. Here, protein kinase D3 (PRKD3) is defined to be a key regulator of CLU in promoting TNBC tumor growth. Mechanically, PRKD3 with kinase activity binding to CLU is critical for CLU protein stability via inhibiting CLU's lysosomal distribution and degradation. CLU and PRKD3 protein level are significantly elevated and positively correlated in collected TNBC tumor samples. CLU silencer (OGX-011) and PRKDs inhibitor (CRT0066101) can both result in impressive tumor growth suppression in vitro and in vivo, suggesting targeting CLU and its key regulator-PRKD3 are promisingly efficient against TNBC. Finally, secreted CLU (sCLU) is found to be elevated in serums from TNBC patients and reduced in serum from TNBC murine models post OGX-011 and/or CRT0066101 treatment, suggesting serum sCLU is a promising blood-based biomarker for clinical management of TNBC. Taken together, this study provides a thorough molecular basis as well as preclinical evidences for targeting CLU pathway as a new promising strategy against TNBC via revealing PRKD3 as the key regulator of CLU in TNBC.
Collapse
Affiliation(s)
- Yan Liu
- The Key Laboratory of Bio‐Medical DiagnosticsSuzhou Institute of Biomedical Engineering and TechnologyChinese Academy of SciencesSuzhou215163P. R. China
- Cancer InstituteDepartment of BiochemistryJiangsu Key Laboratory for Molecular and Medical BiotechnologyCollege of Life ScienceNanjing Normal UniversityNanjing210023P. R. China
| | - Yehui Zhou
- The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhou215006P. R. China
| | - Xinxing Ma
- The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhou215006P. R. China
| | - Liming Chen
- Cancer InstituteDepartment of BiochemistryJiangsu Key Laboratory for Molecular and Medical BiotechnologyCollege of Life ScienceNanjing Normal UniversityNanjing210023P. R. China
| |
Collapse
|
13
|
Praharaj PP, Patra S, Panigrahi DP, Patra SK, Bhutia SK. Clusterin as modulator of carcinogenesis: A potential avenue for targeted cancer therapy. Biochim Biophys Acta Rev Cancer 2020; 1875:188500. [PMID: 33385484 DOI: 10.1016/j.bbcan.2020.188500] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/14/2020] [Accepted: 12/24/2020] [Indexed: 12/30/2022]
Abstract
Clusterin (CLU) is an evolutionary conserved molecular chaperone present in different human tissues and fluids and established to be a significant cancer regulator. It controls several cancer-associated cellular events, including cancer cell proliferation, stemness, survival, metastasis, epithelial-mesenchymal transition, therapy resistance, and inhibition of programmed cell death to support cancer growth and recurrence. This multifunctional role of CLU makes it an ideal target for cancer control. More importantly, genetic and antisense-mediated (OGX-011) inhibition of CLU enhances the anticancer potential of different FDA-approved chemotherapeutic drugs at the clinical level, improving patient's survival. In this review, we have discussed the detailed mechanism of CLU-mediated modulation of different cancer-associated signaling pathways. We have also provided updated information on the current preclinical and clinical findings that drive trials in various cancer types for potential targeted cancer therapy.
Collapse
Affiliation(s)
- Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Debasna Pritimanjari Panigrahi
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| |
Collapse
|
14
|
Naik PP, Mukhopadhyay S, Praharaj PP, Bhol CS, Panigrahi DP, Mahapatra KK, Patra S, Saha S, Panda AK, Panda K, Paul S, Aich P, Patra SK, Bhutia SK. Secretory clusterin promotes oral cancer cell survival via inhibiting apoptosis by activation of autophagy in AMPK/mTOR/ULK1 dependent pathway. Life Sci 2020; 264:118722. [PMID: 33160989 DOI: 10.1016/j.lfs.2020.118722] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/26/2020] [Accepted: 10/31/2020] [Indexed: 02/06/2023]
Abstract
AIMS Secretory clusterin (sCLU) plays an important role in tumor development and cancer progression. However, the molecular mechanisms and physiological functions of sCLU in oral cancer is unclear. We examined the impact of sCLU-mediated autophagy in cell survival and apoptosis inhibition in oral cancer. MAIN METHODS Immunohistochemical analysis was performed to analyze protein expression in patient samples. Autophagy and mitophagy was studied by immunofluorescence microscopy and Western blot. The gain and loss of function was studied by overexpression of plasmid and siRNA approaches respectively. Cellular protection against nutrient starvation and therapeutic stress by sCLU was studied by cell viability, caspase assay and meta-analysis. KEY FINDINGS The data from oral cancer patients showed that the expression levels of sCLU, ATG14, ULK1, and PARKIN increased in grade-wise manners. Interestingly, sCLU overexpression promoted autophagy through AMPK/Akt/mTOR signaling pathway leading to cell survival and protection from long exposure serum starvation induced-apoptosis. Additionally, sCLU was demonstrated to interact with ULK1 and inhibition of ULK1 activity by SBI206965 was found to abolish sCLU-induced autophagy indicating critical role of ULK1 in induction of autophagy. Furthermore, sCLU was observed to promote expression of mitophagy-associated proteins in serum starvation conditions to protect cells from nutrient deprivation. The meta-analysis elucidated that high CLU expression is associated with therapy resistance in cancer and we demonstrated that sCLU-mediated mitophagy was revealed to inhibit cell death by cisplatin. SIGNIFICANCE The present investigation has highlighted the probable implications of the clusterin-induced autophagy in cell survival and inhibition of apoptosis in oral cancer.
Collapse
Affiliation(s)
- Prajna Paramita Naik
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Subhadip Mukhopadhyay
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Chandra Sekhar Bhol
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Debasna Pritimanjari Panigrahi
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Kewal Kumar Mahapatra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Sarbari Saha
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | | | - Krupasindhu Panda
- Panda Curie Cancer Hospital, Telenga Pentha, Cuttack, 753051, Odisha, India
| | - Subhankar Paul
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Palok Aich
- National Institute of Science Education and Research (NISER), HBNI, Bhipmpur-Padanpur, Jatni, Khurda 752050, Odisha, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| |
Collapse
|
15
|
Transcriptomic analysis reveals dynamic molecular changes in skin induced by mechanical forces secondary to tissue expansion. Sci Rep 2020; 10:15991. [PMID: 32994433 PMCID: PMC7524724 DOI: 10.1038/s41598-020-71823-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/07/2020] [Indexed: 12/19/2022] Open
Abstract
Tissue expansion procedures (TE) utilize mechanical forces to induce skin growth and regeneration. While the impact of quick mechanical stimulation on molecular changes in cells has been studied extensively, there is a clear gap in knowledge about sequential biological processes activated during long-term stimulation of skin in vivo. Here, we present the first genome-wide study of transcriptional changes in skin during TE, starting from 1 h to 7 days of expansion. Our results indicate that mechanical forces from a tissue expander induce broad molecular changes in gene expression, and that these changes are time-dependent. We revealed hierarchical changes in skin cell biology, including activation of an immune response, a switch in cell metabolism and processes related to muscle contraction and cytoskeleton organization. In addition to known mechanoresponsive genes (TNC, MMPs), we have identified novel candidate genes (SFRP2, SPP1, CCR1, C2, MSR1, C4A, PLA2G2F, HBB), which might play crucial roles in stretched-induced skin growth. Understanding which biological processes are affected by mechanical forces in TE is important for the development of skin treatments to maximize the efficacy and minimize the risk of complications during expansion procedures.
Collapse
|
16
|
Madden EC, Gorman AM, Logue SE, Samali A. Tumour Cell Secretome in Chemoresistance and Tumour Recurrence. Trends Cancer 2020; 6:489-505. [PMID: 32460003 DOI: 10.1016/j.trecan.2020.02.020] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 12/22/2022]
Abstract
Chemoresistance is a major factor driving tumour relapse and the high rates of cancer-related deaths. Understanding how cancer cells overcome chemotherapy-induced cell death is critical in promoting patient survival. One emerging mechanism of chemoresistance is the tumour cell secretome (TCS), an array of protumorigenic factors released by tumour cells. Chemotherapy exposure can also alter the composition of the TCS, known as therapy-induced TCS, and can promote tumour relapse and the formation of an immunosuppressive tumour microenvironment (TME). Here, we outline how the TCS can protect cancer cells from chemotherapy-induced cell death. We also highlight recent evidence describing how therapy-induced TCS can impact cancer stem cell (CSC) expansion and tumour-associated immune cells to enable tumour regrowth and antitumour immunity.
Collapse
Affiliation(s)
- Emma C Madden
- Apoptosis Research Centre, NUI Galway, Galway, Ireland; School of Natural Sciences, NUI Galway, Galway, Ireland
| | - Adrienne M Gorman
- Apoptosis Research Centre, NUI Galway, Galway, Ireland; School of Natural Sciences, NUI Galway, Galway, Ireland
| | - Susan E Logue
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
| | - Afshin Samali
- Apoptosis Research Centre, NUI Galway, Galway, Ireland; School of Natural Sciences, NUI Galway, Galway, Ireland.
| |
Collapse
|
17
|
Rao CV, Asch AS, Carr DJJ, Yamada HY. "Amyloid-beta accumulation cycle" as a prevention and/or therapy target for Alzheimer's disease. Aging Cell 2020; 19:e13109. [PMID: 31981470 PMCID: PMC7059149 DOI: 10.1111/acel.13109] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/16/2019] [Accepted: 12/25/2019] [Indexed: 02/06/2023] Open
Abstract
The cell cycle and its regulators are validated targets for cancer drugs. Reagents that target cells in a specific cell cycle phase (e.g., antimitotics or DNA synthesis inhibitors/replication stress inducers) have demonstrated success as broad-spectrum anticancer drugs. Cyclin-dependent kinases (CDKs) are drivers of cell cycle transitions. A CDK inhibitor, flavopiridol/alvocidib, is an FDA-approved drug for acute myeloid leukemia. Alzheimer's disease (AD) is another serious issue in contemporary medicine. The cause of AD remains elusive, although a critical role of latent amyloid-beta accumulation has emerged. Existing AD drug research and development targets include amyloid, amyloid metabolism/catabolism, tau, inflammation, cholesterol, the cholinergic system, and other neurotransmitters. However, none have been validated as therapeutically effective targets. Recent reports from AD-omics and preclinical animal models provided data supporting the long-standing notion that cell cycle progression and/or mitosis may be a valid target for AD prevention and/or therapy. This review will summarize the recent developments in AD research: (a) Mitotic re-entry, leading to the "amyloid-beta accumulation cycle," may be a prerequisite for amyloid-beta accumulation and AD pathology development; (b) AD-associated pathogens can cause cell cycle errors; (c) thirteen among 37 human AD genetic risk genes may be functionally involved in the cell cycle and/or mitosis; and (d) preclinical AD mouse models treated with CDK inhibitor showed improvements in cognitive/behavioral symptoms. If the "amyloid-beta accumulation cycle is an AD drug target" concept is proven, repurposing of cancer drugs may emerge as a new, fast-track approach for AD management in the clinic setting.
Collapse
Affiliation(s)
- Chinthalapally V. Rao
- Center for Cancer Prevention and Drug DevelopmentDepartment of MedicineHematology/Oncology SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Adam S. Asch
- Stephenson Cancer CenterDepartment of MedicineHematology/Oncology SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Daniel J. J. Carr
- Department of OphthalmologyUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Hiroshi Y. Yamada
- Center for Cancer Prevention and Drug DevelopmentDepartment of MedicineHematology/Oncology SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| |
Collapse
|
18
|
Dong Z, Jiao Y, Xie B, Hao Y, Wang P, Liu Y, Shi J, Chitrakar C, Black S, Wang YC, Lee LJ, Li M, Fan Y, Chang L. On-chip multiplexed single-cell patterning and controllable intracellular delivery. MICROSYSTEMS & NANOENGINEERING 2020; 6:2. [PMID: 34567617 PMCID: PMC8433345 DOI: 10.1038/s41378-019-0112-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/23/2019] [Accepted: 10/10/2019] [Indexed: 05/03/2023]
Abstract
Conventional electroporation approaches show limitations in the delivery of macromolecules in vitro and in vivo. These limitations include low efficiency, noticeable cell damage and nonuniform delivery of cells. Here, we present a simple 3D electroporation platform that enables massively parallel single-cell manipulation and the intracellular delivery of macromolecules and small molecules. A pyramid pit micropore array chip was fabricated based on a silicon wet-etching method. A controllable vacuum system was adopted to trap a single cell on each micropore. Using this chip, safe single-cell electroporation was performed at low voltage. Cargoes of various sizes ranging from oligonucleotides (molecular beacons, 22 bp) to plasmid DNA (CRISPR-Cas9 expression vectors, >9 kb) were delivered into targeted cells with a significantly higher transfection efficiency than that of multiple benchmark methods (e.g., commercial electroporation devices and Lipofectamine). The delivered dose of the chemotherapeutic drug could be controlled by adjusting the applied voltage. By using CRISPR-Cas9 transfection with this system, the p62 gene and CXCR7 gene were knocked out in tumor cells, which effectively inhibited their cellular activity. Overall, this vacuum-assisted micropore array platform provides a simple, efficient, high-throughput intracellular delivery method that may facilitate on-chip cell manipulation, intracellular investigation and cancer therapy.
Collapse
Affiliation(s)
- Zaizai Dong
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Yanli Jiao
- College of Agricultural and Life Sciences, University of Florida, Gainesville, FL 32611 USA
| | - Bingteng Xie
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yongcun Hao
- Ministry of Education Key Laboratory of Micro and Nano Systems for Aerospace, Northwestern Polytechnical University, 710072 Xi’an, China
| | - Pan Wang
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yuanyuan Liu
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Junfeng Shi
- Chemical and Biomolecular Engineering Department, Ohio State University, Columbus, OH 43209 USA
| | - Chandani Chitrakar
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| | - Stephen Black
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| | - Yu-Chieh Wang
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| | - L. James Lee
- Chemical and Biomolecular Engineering Department, Ohio State University, Columbus, OH 43209 USA
| | - Mo Li
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yubo Fan
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Lingqian Chang
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| |
Collapse
|
19
|
Bertacchini J, Mediani L, Beretti F, Guida M, Ghalali A, Brugnoli F, Bertagnolo V, Petricoin E, Poti F, Arioli J, Anselmi L, Bari A, McCubrey J, Martelli AM, Cocco L, Capitani S, Marmiroli S. Clusterin enhances AKT2-mediated motility of normal and cancer prostate cells through a PTEN and PHLPP1 circuit. J Cell Physiol 2019; 234:11188-11199. [PMID: 30565691 DOI: 10.1002/jcp.27768] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 10/30/2018] [Indexed: 07/23/2024]
Abstract
Clusterin (CLU) is a chaperone-like protein with multiple functions. sCLU is frequently upregulated in prostate tumor cells after chemo- or radiotherapy and after surgical or pharmacological castration. Moreover, CLU has been documented to modulate the cellular homolog of murine thymoma virus akt8 oncogene (AKT) activity. Here, we investigated how CLU overexpression influences phosphatidylinositol 3'-kinase (PI3K)/AKT signaling in human normal and cancer epithelial prostate cells. Human prostate cells stably transfected with CLU were broadly profiled by reverse phase protein array (RPPA), with particular emphasis on the PI3K/AKT pathway. The effect of CLU overexpression on normal and cancer cell motility was also tested. Our results clearly indicate that CLU overexpression enhances phosphorylation of AKT restricted to isoform 2. Mechanistically, this can be explained by the finding that the phosphatase PH domain leucine-rich repeat-containing protein phosphatase 1 (PHLPP1), known to dephosphorylate AKT2 at S474, is markedly downregulated by CLU, whereas miR-190, a negative regulator of PHLPP1, is upregulated. Moreover, we found that phosphatase and tensin homolog (PTEN) was heavily phosphorylated at the inhibitory site S380, contributing to the hyperactivation of AKT signaling. By keeping AKT2 phosphorylation high, CLU dramatically enhances the migratory behavior of prostate epithelial cell lines with different migratory and invasive phenotypes, namely prostate normal epithelial 1A (PNT1A) and prostatic carcinoma 3 (PC3) cells. Altogether, our results unravel for the first time a circuit by which CLU can switch a low migration phenotype toward a high migration phenotype, through miR-190-dependent downmodulation of PHLPP1 expression and, in turn, stabilization of AKT2 phosphorylation.
Collapse
Affiliation(s)
- Jessika Bertacchini
- Department of Biomedical, Metabolic, and Neural Sciences, Section of Morphology, Signal Transduction Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Mediani
- Department of Biomedical, Metabolic, and Neural Sciences, Section of Morphology, Signal Transduction Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Beretti
- Department of Medicine, Surgery, Dentistry, and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Marianna Guida
- Department of Biomedical, Metabolic, and Neural Sciences, Section of Morphology, Signal Transduction Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Aram Ghalali
- Institute of Environment Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Federica Brugnoli
- Department of Morphology, Surgery, and Experimental Medicine, Section of Anatomy and Histology and LTTA Center, University of Ferrara, Ferrara, Italy
| | - Valeria Bertagnolo
- Department of Morphology, Surgery, and Experimental Medicine, Section of Anatomy and Histology and LTTA Center, University of Ferrara, Ferrara, Italy
| | - Emanuel Petricoin
- Center for Applied Proteomics & Molecular Medicine, GMU, Fairfax, Virginia
| | - Francesco Poti
- Department of Medicine and Surgery-Unit of Neurosciences, University of Parma, Parma, Italy
| | - Jessica Arioli
- Department of Biomedical, Metabolic, and Neural Sciences, Section of Morphology, Signal Transduction Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Anselmi
- Department of Biomedical, Metabolic, and Neural Sciences, Section of Morphology, Signal Transduction Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessia Bari
- Department of Diagnostic, Clinical Medicine and Public Health, Program of Innovative Therapy in Oncology and Hematology, University of Modena and Reggio Emilia, Modena, Italy
| | - James McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | - Alberto M Martelli
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Silvano Capitani
- Department of Morphology, Surgery, and Experimental Medicine, Section of Anatomy and Histology and LTTA Center, University of Ferrara, Ferrara, Italy
| | - Sandra Marmiroli
- Department of Biomedical, Metabolic, and Neural Sciences, Section of Morphology, Signal Transduction Unit, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
20
|
Liu R, Ma Z, Kunle Kolawole S, Zeng L, Zhao Y, Ren L, Yang K. In vitro study on cytocompatibility and osteogenesis ability of Ti-Cu alloy. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2019; 30:75. [PMID: 31218519 DOI: 10.1007/s10856-019-6277-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 06/03/2019] [Indexed: 06/09/2023]
Abstract
Titanium implants easily suffer bacteria-related infections in clinic due to their inherent lack of self-protection ability. Therefore, a novel Ti-Cu alloy with good antibacterial activity has been developed as a new kind of implant material. This study focuses on a systematic evaluation of both cytocompatibility and osteogenesis activity of the Ti-Cu alloy in vitro. It was revealed that an addition of 5% Cu into pure Ti would not cause any negative effect on osteoblasts adhesion, proliferation and apoptosis cultured with Ti-Cu alloy. In addition, Ti-Cu alloy could significantly promote the osteogenic differentiation of MG 63 cells by upregulating the osteogenesis-related gene expressions including alkaline phosphatase (ALP), Collagen I (Colla I), osteopontin (OPN) and osteocalcin (OCN). These promising results suggest that the Ti-Cu alloy has great potential to be used as a multi-functional titanium implant for clinical applications.
Collapse
Affiliation(s)
- Rui Liu
- Northeastern University, 3-11 Wenhua Road, 110819, Shenyang, China
- Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, 110016, Shenyang, China
| | - Zheng Ma
- Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, 110016, Shenyang, China
| | | | - Lilan Zeng
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, 518055, Shenzhen, China
| | - Ying Zhao
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, 518055, Shenzhen, China.
| | - Ling Ren
- Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, 110016, Shenyang, China.
| | - Ke Yang
- Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, 110016, Shenyang, China.
| |
Collapse
|
21
|
Bouaouiche S, Magadoux L, Dondaine L, Reveneau S, Isambert N, Bettaieb A, Jeannin JF, Laurens V, Plenchette S. Glyceryl trinitrate‑induced cytotoxicity of docetaxel‑resistant prostatic cancer cells is associated with differential regulation of clusterin. Int J Oncol 2019; 54:1446-1456. [PMID: 30720069 DOI: 10.3892/ijo.2019.4708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/21/2018] [Indexed: 11/05/2022] Open
Abstract
Metastatic castration resistant prostate cancer (mCRPC) relapse due to acquired resistance to chemotherapy, such as docetaxel, remains a major threat to patient survival. Resistance of mCRPC to docetaxel can be associated with elevated levels of soluble clusterin (sCLU) and growth differentiation factor‑15 (GDF‑15). Any strategies aiming to modulate sCLU and/or GDF‑15 in docetaxel‑resistant prostate cancer cells present a therapeutic interest. The present study reports the cytotoxic effect of a nitric oxide donor, glyceryl trinitrate (GTN), on docetaxel‑resistant mCRPC human cell lines and demonstrates that GTN displays greater inhibition of cell viability toward docetaxel‑resistant mCRPC cells than on mCRPC cells. It is also demonstrated that GTN modulates the level of expression of clusterin (CLU) which is dependent of GDF‑15, two markers associated with docetaxel resistance in prostate cancer. The results indicate that GTN represses the level of expression of the cytoprotective isoform of CLU (sCLU) and can increase the level of expression of the cytotoxic isoform (nuclear CLU) in docetaxel resistant cells. Furthermore, it was observed that GTN differentially regulates the level of the precursor form of GDF‑15 between resistant and parental cells, and that recombinant GDF‑15 can modulate the expression of CLU isoforms and counteract GTN‑induced cytotoxicity in resistant cells. A link was established between GDF‑15 and the expression of CLU isoforms. The present study thus revealed GTN as a potential therapeutic strategy to overcome docetaxel‑resistant mCRPC.
Collapse
Affiliation(s)
- Sarra Bouaouiche
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Lea Magadoux
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Lucile Dondaine
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Sylvie Reveneau
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | | | - Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Jean-François Jeannin
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Veronique Laurens
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Stephanie Plenchette
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| |
Collapse
|
22
|
Liu X, Chen Y, Li Y, Petersen RB, Huang K. Targeting mitosis exit: A brake for cancer cell proliferation. Biochim Biophys Acta Rev Cancer 2019; 1871:179-191. [PMID: 30611728 DOI: 10.1016/j.bbcan.2018.12.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 12/16/2022]
Abstract
The transition from mitosis to interphase, referred to as mitotic exit, is a critical mitotic process which involves activation and inactivation of multiple mitotic kinases and counteracting protein phosphatases. Loss of mitotic exit checkpoints is a common feature of cancer cells, leading to mitotic dysregulation and confers cancer cells with oncogenic characteristics, such as aberrant proliferation and microtubule-targeting agent (MTA) resistance. Since MTA resistance results from cancer cells prematurely exiting mitosis (mitotic slippage), blocking mitotic exit is believed to be a promising anticancer strategy. Moreover, based on this theory, simultaneous inhibition of mitotic exit and additional cell cycle phases would likely achieve synergistic antitumor effects. In this review, we divide the molecular regulators of mitotic exit into four categories based on their different regulatory functions: 1) the anaphase-promoting complex/cyclosome (APC/C, a ubiquitin ligase), 2) cyclin B, 3) mitotic kinases and phosphatases, 4) kinesins and microtubule-binding proteins. We also review the regulators of mitotic exit and propose prospective anticancer strategies targeting mitotic exit, including their strengths and possible challenges to their use.
Collapse
Affiliation(s)
- Xinran Liu
- Tongji School of Pharmacy, Huazhong University of Science & Technology, Wuhan, Hubei 430030, China
| | - Yuchen Chen
- Tongji School of Pharmacy, Huazhong University of Science & Technology, Wuhan, Hubei 430030, China
| | - Yangkai Li
- Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Robert B Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, MI 48858, USA
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science & Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
23
|
Xiong J, Wang S, Chen T, Shu X, Mo X, Chang G, Chen JJ, Li C, Luo H, Lee JD. Verteporfin blocks Clusterin which is required for survival of gastric cancer stem cell by modulating HSP90 function. Int J Biol Sci 2019; 15:312-324. [PMID: 30745823 PMCID: PMC6367548 DOI: 10.7150/ijbs.29135] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/11/2018] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer stem cell (GCSC) is implicated in gastric cancer relapse, metastasis and drug resistance. However, the key molecule(s) involved in GCSC survival and the targeting drugs are poorly understood. We discovered increased secreted clusterin (S-Clu) protein expression during the sphere-forming growth of GCSC via mass spectrometry. Overexpression of clusterin was detected in 69/90 (77%) of primary GC tissues and significantly associated with T stage, lymph node metastasis and TNM stage. Depletion of clusterin (Clu, the full-length intracellular clusterin) led to the declustering of GCSC tumorspheres and apoptosis of GCSC. Subsequently, we found clusterin was in complex with heat shock protein 90 beta (HSP90) and involved in regulating the cellular level of HSP90 client proteins. Furthermore, by screening a collection of drugs/inhibitors, we found that verteporfin (VP), a phototherapy drug, blocked clusterin gene expression, decreased the HSP90 client proteins and caused cell death of GCSC. VP treatment is more effective in eradicating GCSCs than in killing GC cells. Both clusterin silencing or VP treatment deterred tumor growth in human GCSC xenografts. These findings collectively suggest that GC patients can promptly benefit from clusterin-targeted therapy as well as VP treatment in combination with or subsequent to conventional chemotherapy for reducing mortality of GC.
Collapse
Affiliation(s)
- Jixian Xiong
- School of Medicine, Shenzhen University, Shenzhen 518055, China.,Shenzhen University International Cancer Center, Shenzhen University, Shenzhen 518055, China
| | - Shaoxiang Wang
- School of Medicine, Shenzhen University, Shenzhen 518055, China
| | - Tie Chen
- School of Medicine, Shenzhen University, Shenzhen 518055, China
| | - Xingsheng Shu
- School of Medicine, Shenzhen University, Shenzhen 518055, China
| | - Xianming Mo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gang Chang
- School of Medicine, Shenzhen University, Shenzhen 518055, China
| | - Jia-Jie Chen
- School of Medicine, Shenzhen University, Shenzhen 518055, China
| | - Chenyang Li
- School of Medicine, Shenzhen University, Shenzhen 518055, China
| | - Hui Luo
- School of Medicine, Shenzhen University, Shenzhen 518055, China
| | - Jiing-Dwan Lee
- School of Medicine, Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
24
|
Purushothaman B, Arumugam P, Ju H, Kulsi G, Samson AAS, Song JM. Novel ruthenium(II) triazine complex [Ru(bdpta)(tpy)]2+ co-targeting drug resistant GRP78 and subcellular organelles in cancer stem cells. Eur J Med Chem 2018; 156:747-759. [DOI: 10.1016/j.ejmech.2018.07.048] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/14/2018] [Accepted: 07/17/2018] [Indexed: 12/17/2022]
|
25
|
Chi KN, Higano CS, Blumenstein B, Ferrero JM, Reeves J, Feyerabend S, Gravis G, Merseburger AS, Stenzl A, Bergman AM, Mukherjee SD, Zalewski P, Saad F, Jacobs C, Gleave M, de Bono JS. Custirsen in combination with docetaxel and prednisone for patients with metastatic castration-resistant prostate cancer (SYNERGY trial): a phase 3, multicentre, open-label, randomised trial. Lancet Oncol 2017; 18:473-485. [PMID: 28283282 DOI: 10.1016/s1470-2045(17)30168-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/17/2017] [Accepted: 01/20/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Clusterin is a chaperone protein associated with treatment resistance and upregulated by apoptotic stressors such as chemotherapy. Custirsen is a second-generation antisense oligonucleotide that inhibits clusterin production. The aim of the SYNERGY trial was to investigate the effect of custirsen in combination with docetaxel and prednisone on overall survival in patients with metastatic castration-resistant prostate cancer. METHODS SYNERGY was a phase 3, multicentre, open-label, randomised trial set at 134 study centres in 12 countries. Patients were eligible for participation if they had: metastatic castration-resistant prostate cancer and had received no previous chemotherapy; prostate-specific antigen greater than 5 ng/mL; and a Karnofsky performance score of 70% or higher. Patients were randomly assigned 1:1 centrally to either the docetaxel, prednisone, and custirsen combination or docetaxel and prednisone alone. Patients were not masked to treatment allocation. Randomisation was stratified by opioid use for cancer-related pain and radiographic evidence of progression. All patients received docetaxel 75 mg/m2 intravenously with 5 mg of prednisone orally twice daily. Patients assigned docetaxel, prednisone, and custirsen received weekly doses of custirsen 640 mg intravenously after three loading doses of 640 mg. The primary endpoint was overall survival analysed in the intention-to-treat population. Patients who received at least one study dose were included in the safety analysis set. This trial is registered with ClinicalTrials.gov, number NCT01188187. The trial is completed and final analyses are reported here. FINDINGS Between Dec 10, 2010, and Nov 7, 2012, 1022 patients were enrolled to the trial, of whom 510 were assigned docetaxel, prednisone, and custirsen and 512 were allocated docetaxel and prednisone. No difference in overall survival was recorded between the two groups (median survival 23·4 months [95% CI 20·9-24·8] with docetaxel, prednisone, and custirsen vs 22·0 months [19·5-24·0] with docetaxel and prednisone; hazard ratio [HR] 0·93, 95% CI 0·79-1·10; p=0·415). The most common adverse events of grade 3 or worse in the docetaxel, prednisone and custirsen group (n=501) compared with the docetaxel and prednisone alone group (n=499) were neutropenia (grade 3, 63 [13%] vs 28 [6%]; grade 4, 98 [20%] vs 77 [15%]), febrile neutropenia (grade 3, 52 [10%] vs 31 [6%]; grade 4, four [1%] vs two [<1%]), and fatigue (grade 3, 53 [11%] vs 41 [8%]; grade 4, three [1%] vs one [<1%]). One or more serious adverse events were reported for 214 (43%) of 501 patients treated with docetaxel, prednisone, and custirsen and 181 (36%) of 499 receiving docetaxel and prednisone alone. Adverse events were attributable to 23 (5%) deaths in the docetaxel, prednisone, and custirsen group and 24 (5%) deaths in the docetaxel and prednisone alone group. INTERPRETATION Addition of custirsen to first-line docetaxel and prednisone was reasonably well tolerated, but overall survival was not significantly longer for patients with metastatic castration-resistant prostate cancer treated with this combination, compared with patients treated with docetaxel and prednisone alone. FUNDING OncoGenex Technologies.
Collapse
Affiliation(s)
- Kim N Chi
- Department of Medicine, Division of Medical Oncology, University of British Columbia, BC Cancer Agency, Vancouver Prostate Centre, Vancouver, BC, Canada.
| | - Celestia S Higano
- Department of Medicine, Division of Medical Oncology, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | - Gwenaelle Gravis
- Institut Paoli Calmette, Department of Cancer Medicine, Marseille, France
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | | | - Andries M Bergman
- Netherlands Cancer Institute and Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Som D Mukherjee
- Juravinski Cancer Centre at Hamilton Health Sciences, Hamilton, ON, Canada
| | | | - Fred Saad
- Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | | | - Martin Gleave
- Department of Medicine, Division of Medical Oncology, University of British Columbia, BC Cancer Agency, Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Johann S de Bono
- Drug Development Unit, Royal Marsden Hospital and Institute of Cancer Research, London, UK
| |
Collapse
|
26
|
Hotte SJ. Addressing taxane resistance in metastatic castration-resistant prostate cancer: a focus on chaperone proteins. Future Oncol 2017; 13:369-379. [DOI: 10.2217/fon-2016-0279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Despite the significant survival benefit of taxane therapy in metastatic castration-resistant prostate cancer (mCRPC), all patients inevitably develop treatment resistance. An understanding of resistance mechanisms has led to new therapies for prostate cancer (cabazitaxel, abiraterone and enzalutamide), all of which have improved survival following first-line docetaxel. Another treatment, currently in development, targets the prosurvival molecule clusterin. Custirsen, an antisense molecule that inhibits clusterin production, has shown promise in combination with docetaxel in mCRPC patients at risk for poor outcomes. Although optimal sequence and combination of available therapies is unclear, the heterogeneity of mCRPC suggests a continuing need for personalized treatment regimens and improved abilities to predict which patients will respond to the available treatment options.
Collapse
Affiliation(s)
- Sebastien J Hotte
- Department of Oncology, Division of Medical Oncology, Juravinski Cancer Centre, 699 Concession Street, Hamilton, Ontario, L8V 5C2, Canada
| |
Collapse
|
27
|
Knockdown of clusterin alters mitochondrial dynamics, facilitates necrosis in camptothecin-induced cancer stem cells. Cell Biol Toxicol 2017; 33:307-321. [DOI: 10.1007/s10565-016-9378-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/22/2016] [Indexed: 10/20/2022]
|
28
|
Abstract
INTRODUCTION Clusterin (CLU) is a stress-activated, ATP-independent molecular chaperone, normally secreted from cells, that is up-regulated in Alzheimer disease and in many cancers. It plays important roles in protein homeostasis/proteostasis, inhibition of cell death pathways, and modulation of pro-survival signalling and transcriptional networks. Changes in the CLU gene locus are highly associated with Alzheimer disease, and many therapy-resistant cancers over-express CLU. The extensive post-translational processing and heterogeneous oligomerization of CLU have so far prevented any definitive structure determination. This in turn has meant that targeting CLU with small molecule inhibitors is challenging. Therefore, inhibiting CLU at the gene-expression level using siRNA or antisense is a valid approach to inhibit its function. Areas covered: This article reviews recent advances regarding the role of CLU in proteostasis, cellular trafficking, human diseases, and signalling pathways involved in oncogenesis. It addresses the rationale for CLU as a therapeutic target in cancer, and the current status of pre-clinical and clinical studies using CLU antisense inhibitor OGX011. Expert opinion: Discusses challenges facing the therapeutic targeting of CLU including rapid changes in the treatment landscape for prostate cancer with multiple new FDA approved drugs, selection of windows of intervention, and potential side effects when silencing CLU expression.
Collapse
Affiliation(s)
- Mark R Wilson
- a School of Biological Sciences , University of Wollongong , Wollongong , Australia
| | - Amina Zoubeidi
- b Department of Urologic Sciences, Vancouver Prostate Centre , University of British Columbia and Vancouver General Hospital , Vancouver , Canada
| |
Collapse
|
29
|
Al Nakouzi N, Wang CK, Beraldi E, Jager W, Ettinger S, Fazli L, Nappi L, Bishop J, Zhang F, Chauchereau A, Loriot Y, Gleave M. Clusterin knockdown sensitizes prostate cancer cells to taxane by modulating mitosis. EMBO Mol Med 2016; 8:761-78. [PMID: 27198502 PMCID: PMC4931290 DOI: 10.15252/emmm.201506059] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Clusterin (CLU) is a stress‐activated molecular chaperone that confers treatment resistance to taxanes when highly expressed. While CLU inhibition potentiates activity of taxanes and other anti‐cancer therapies in preclinical models, progression to treatment‐resistant disease still occurs implicating additional compensatory survival mechanisms. Taxanes are believed to selectively target cells in mitosis, a complex mechanism controlled in part by balancing antagonistic roles of Cdc25C and Wee1 in mitosis progression. Our data indicate that CLU silencing induces a constitutive activation of Cdc25C, which delays mitotic exit and hence sensitizes cancer cells to mitotic‐targeting agents such as taxanes. Unchecked Cdc25C activation leads to mitotic catastrophe and cell death unless cells up‐regulate protective mechanisms mediated through the cell cycle regulators Wee1 and Cdk1. In this study, we show that CLU silencing induces a constitutive activation of Cdc25C via the phosphatase PP2A leading to relief of negative feedback inhibition and activation of Wee1‐Cdk1 to promote survival and limit therapeutic efficacy. Simultaneous inhibition of CLU‐regulated cell cycle effector Wee1 may improve synergistic responses of biologically rational combinatorial regimens using taxanes and CLU inhibitors.
Collapse
Affiliation(s)
- Nader Al Nakouzi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Chris Kedong Wang
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Eliana Beraldi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Wolfgang Jager
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Susan Ettinger
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ladan Fazli
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Lucia Nappi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jennifer Bishop
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Fan Zhang
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Anne Chauchereau
- Department of Cancer Medicine, Gustave Roussy, Cancer Campus, Grand Paris, University of Paris-Sud, Villejuif, France INSERM U981, Villejuif, France
| | - Yohann Loriot
- Department of Cancer Medicine, Gustave Roussy, Cancer Campus, Grand Paris, University of Paris-Sud, Villejuif, France INSERM U981, Villejuif, France
| | - Martin Gleave
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|