1
|
Pallathadka H, Jabir M, Rasool KH, Hanumanthaiah M, Sharma N, Pramanik A, Rab SO, Jawad SF, Oghenemaro EF, Mustafa YF. siRNA-based therapy for overcoming drug resistance in human solid tumours; molecular and immunological approaches. Hum Immunol 2025; 86:111221. [PMID: 39700968 DOI: 10.1016/j.humimm.2024.111221] [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: 08/10/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
RNA interference (RNAi) is a primordial biological process that protects against external intrusion. SiRNA has the potential to selectively silence disease-related genes in a sequence-specific way, thus offering a promising therapeutic approach. The efficacy of siRNA-based therapies in cancer treatment has gained significant recognition due to multiple studies demonstrating its ability to effectively suppress cancer cells' growth and multiplication. Moreover, siRNA-based medicines have shown considerable promise in enhancing the sensitivity of cancer cells to chemotherapy and other treatment methods by suppressing genes that play a role in the development of drug resistance. Exploring and identifying functional genes linked to cancer cell characteristics and drug resistance is crucial for developing effective siRNAs for cancer treatment and advancing targeted and personalized therapeutics. Targeting and silencing genes in charge of resistance mechanisms, such as those involved in drug efflux, cell survival, or DNA repair, is possible with siRNA therapy in the context of drug resistance, especially cancer. Through inhibiting these genes, siRNA therapy can prevent resistance and restore the efficacy of traditional medications. This review addresses the potential of siRNAs in addressing drug resistance in human tumours, opening up new possibilities in cancer therapy. This review article offers a non-systematic summary of how different siRNA types contribute to cancer cells' treatment resistance. Using pertinent keywords, sources were chosen from reliable databases, including PubMed, Scopus, and Google Scholar. The review covered essential papers in this area and those that mainly addressed the function of siRNA in drug resistance. The articles examined in connection with the title of this review were primarily published from 2020 onward and are based on in vitro studies. Furthermore, this article examines the potential barriers and prospective perspectives of siRNA therapies.
Collapse
Affiliation(s)
| | - Majid Jabir
- Department of Applied Sciences, University of Technology, Iraq
| | | | - Malathi Hanumanthaiah
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Neha Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri - 140307, Mohali, Punjab, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University Dehradun, Uttarakhand, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Sabrean Farhan Jawad
- Department of Biochemistry, College of Science, Al-Mustaqbal University, 51001 Babil, Iraq.
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Delta State University, Faculty of Pharmacy, PMB 1 Abraka, Delta State, Nigeria
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
2
|
Wesley T, Escalona RM, Kannourakis G, Ahmed N. Plakin Expression in Serous Epithelial Ovarian Cancer Has the Potential to Impede Metastatic Spread and Epithelial-Mesenchymal Transition: A Comparative Expression Analysis of Immunohistochemical and In Silico Datasets. Cancers (Basel) 2024; 16:4087. [PMID: 39682273 DOI: 10.3390/cancers16234087] [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: 10/23/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Epithelial ovarian cancer is aggressive and causes high mortality among women worldwide. Members of the plakin family are essential to maintain cytoskeletal integrity and key cellular processes. In this study we characterised the expression of plakins, particularly plectin (PLEC), periplakin (PPL), envoplakin (EVPL), and EMT-related proteins by immunohistochemistry in n = 48 patients' samples to evaluate a potential correlation of plakin expression with EMT as EOC progresses. These tissue plakin and EMT expression analyses were further evaluated by in vitro cell line expression and correlated with the expression of these molecules using publicly available datasets such as Cancer Genome Atlas (TCGA) and Clinical Proteome Tumour Analysis Consortium (CPTAC) datasets. We demonstrate that the expression of PPL and PLEC plakins is decreased in high-grade compared to low-grade EOCs with mixed EMT marker protein expression. This is supported by the correlation of high PPL and PLEC expression with an epithelial rather than mesenchymal phenotype. Our data suggest a partial loss of plakin expression as EOC tumours progress. This may impact the connections of plakins with membrane-bound receptors, which impede the downstream signalling required for the initiation of EMT as the tumours progress.
Collapse
Affiliation(s)
- Tamsin Wesley
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Health Innovation and Transformation Centre, Mt Helen Campus, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Ruth M Escalona
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Health Innovation and Transformation Centre, Mt Helen Campus, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Nuzhat Ahmed
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Health Innovation and Transformation Centre, Mt Helen Campus, Federation University Australia, Ballarat, VIC 3353, Australia
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular & Translational Science, Monash University, Clayton, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3050, Australia
- Department of Surgery, St Vincent Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| |
Collapse
|
3
|
Shiau C, Cao J, Gong D, Gregory MT, Caldwell NJ, Yin X, Cho JW, Wang PL, Su J, Wang S, Reeves JW, Kim TK, Kim Y, Guo JA, Lester NA, Bae JW, Zhao R, Schurman N, Barth JL, Ganci ML, Weissleder R, Jacks T, Qadan M, Hong TS, Wo JY, Roberts H, Beechem JM, Castillo CFD, Mino-Kenudson M, Ting DT, Hemberg M, Hwang WL. Spatially resolved analysis of pancreatic cancer identifies therapy-associated remodeling of the tumor microenvironment. Nat Genet 2024; 56:2466-2478. [PMID: 39227743 PMCID: PMC11816915 DOI: 10.1038/s41588-024-01890-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/30/2024] [Indexed: 09/05/2024]
Abstract
In combination with cell-intrinsic properties, interactions in the tumor microenvironment modulate therapeutic response. We leveraged single-cell spatial transcriptomics to dissect the remodeling of multicellular neighborhoods and cell-cell interactions in human pancreatic cancer associated with neoadjuvant chemotherapy and radiotherapy. We developed spatially constrained optimal transport interaction analysis (SCOTIA), an optimal transport model with a cost function that includes both spatial distance and ligand-receptor gene expression. Our results uncovered a marked change in ligand-receptor interactions between cancer-associated fibroblasts and malignant cells in response to treatment, which was supported by orthogonal datasets, including an ex vivo tumoroid coculture system. We identified enrichment in interleukin-6 family signaling that functionally confers resistance to chemotherapy. Overall, this study demonstrates that characterization of the tumor microenvironment using single-cell spatial transcriptomics allows for the identification of molecular interactions that may play a role in the emergence of therapeutic resistance and offers a spatially based analysis framework that can be broadly applied to other contexts.
Collapse
Affiliation(s)
- Carina Shiau
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jingyi Cao
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dennis Gong
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT Health Sciences and Technology Program, Cambridge, MA, USA
| | | | - Nicholas J Caldwell
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xunqin Yin
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jae-Won Cho
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter L Wang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jennifer Su
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Steven Wang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | | - Jimmy A Guo
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Biological and Biomedical Sciences Program, Harvard Medical School, Boston, MA, USA
| | - Nicole A Lester
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jung Woo Bae
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ryan Zhao
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Jamie L Barth
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria L Ganci
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tyler Jacks
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Motaz Qadan
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jennifer Y Wo
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hannah Roberts
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David T Ting
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martin Hemberg
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - William L Hwang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
4
|
Chu X, Tian W, Ning J, Xiao G, Zhou Y, Wang Z, Zhai Z, Tanzhu G, Yang J, Zhou R. Cancer stem cells: advances in knowledge and implications for cancer therapy. Signal Transduct Target Ther 2024; 9:170. [PMID: 38965243 PMCID: PMC11224386 DOI: 10.1038/s41392-024-01851-y] [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: 10/02/2023] [Revised: 03/27/2024] [Accepted: 04/28/2024] [Indexed: 07/06/2024] Open
Abstract
Cancer stem cells (CSCs), a small subset of cells in tumors that are characterized by self-renewal and continuous proliferation, lead to tumorigenesis, metastasis, and maintain tumor heterogeneity. Cancer continues to be a significant global disease burden. In the past, surgery, radiotherapy, and chemotherapy were the main cancer treatments. The technology of cancer treatments continues to develop and advance, and the emergence of targeted therapy, and immunotherapy provides more options for patients to a certain extent. However, the limitations of efficacy and treatment resistance are still inevitable. Our review begins with a brief introduction of the historical discoveries, original hypotheses, and pathways that regulate CSCs, such as WNT/β-Catenin, hedgehog, Notch, NF-κB, JAK/STAT, TGF-β, PI3K/AKT, PPAR pathway, and their crosstalk. We focus on the role of CSCs in various therapeutic outcomes and resistance, including how the treatments affect the content of CSCs and the alteration of related molecules, CSCs-mediated therapeutic resistance, and the clinical value of targeting CSCs in patients with refractory, progressed or advanced tumors. In summary, CSCs affect therapeutic efficacy, and the treatment method of targeting CSCs is still difficult to determine. Clarifying regulatory mechanisms and targeting biomarkers of CSCs is currently the mainstream idea.
Collapse
Affiliation(s)
- Xianjing Chu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wentao Tian
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jiaoyang Ning
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Gang Xiao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yunqi Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziqi Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhuofan Zhai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Guilong Tanzhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jie Yang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| |
Collapse
|
5
|
Ravindran F, Mhatre A, Koroth J, Narayan S, Choudhary B. Curcumin modulates cell type-specific miRNA networks to induce cytotoxicity in ovarian cancer cells. Life Sci 2023; 334:122224. [PMID: 38084671 DOI: 10.1016/j.lfs.2023.122224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 12/18/2023]
Abstract
AIM To understand the epigenetic role of curcumin, a natural polyphenolic compound extracted from the spice Curcuma longa in inducing cytotoxicity in two molecularly distinct ovarian cancer cell lines: PA1 and A2780. MATERIALS AND METHODS An integrated mRNA-miRNA sequence analysis was performed to determine the curcumin-induced mRNA-miRNA regulatory networks in the induction of cytotoxicity. The miRNA-mRNA pathways, the miRNAs and their targets implicated in apoptosis, autophagy, DNA damage, and stemness markers were validated. Gene/miRNA expressions were validated using qPCR and protein expressions by western blotting. Curcumin-induced oncogenic /tumor-suppressor miRNAs were profiled utilising the oncomiRdb database. Similarly, the expressions of oncogenes/tumor suppressor genes were profiled and correlated with the TCGA ovarian cancer dataset. A dual luciferase assay was performed to investigate the interaction of miR-199a-5p to its direct target, DDR1. KEY FINDINGS The expression of several miRNAs demonstrated an inverse correlation with their respective direct targets. In curcumin-treated PA1 cells, miR-335-5p target ATG5 (autophagic), and OCT4 (pluripotent gene) were downregulated, miR-32a target PTEN (tumor suppressor) was upregulated, miR-1285 target P53 (tumor suppressor) was upregulated, and both miR-182-5p and miR-503-3p target BCL2, were down-regulated. Contrastingly, in curcumin-treated A2780 cells, miR-181a-3p target ATG5, miR-30a-5p, and miR-216a target BECN1 (autophagic) were upregulated, and miR-129a-5p target BCL2 were downregulated. The reversal of the oncomiR/TSmiR profile revealed suppression of oncogenic processes by curcumin. Curcumin treatment induced a moderate cisplatin-sensitisation effect and impaired epithelial-to-mesenchymal transition (EMT) characteristics. Curcumin also regulated the miR-199a-5p/DDR1 axis with a decrease in collagen deposition. SIGNIFICANCE The activity of curcumin is cell-type specific. Distinct miRNA regulatory networks were activated to induce multiple modes of cellular cytotoxicity in these ovarian cancer cells. This study further highlights the molecular mechanism of curcumin action in ovarian cancers establishing its candidacy as a promising drug candidate.
Collapse
Affiliation(s)
- Febina Ravindran
- Institute of Bioinformatics and Applied Biotechnology, Electronic city phase 1, Bangalore, India
| | - Anisha Mhatre
- Institute of Bioinformatics and Applied Biotechnology, Electronic city phase 1, Bangalore, India
| | - Jinsha Koroth
- Institute of Bioinformatics and Applied Biotechnology, Electronic city phase 1, Bangalore, India
| | - Suchitra Narayan
- Institute of Bioinformatics and Applied Biotechnology, Electronic city phase 1, Bangalore, India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology, Electronic city phase 1, Bangalore, India.
| |
Collapse
|
6
|
Shiau C, Cao J, Gregory MT, Gong D, Yin X, Cho JW, Wang PL, Su J, Wang S, Reeves JW, Kim TK, Kim Y, Guo JA, Lester NA, Schurman N, Barth JL, Weissleder R, Jacks T, Qadan M, Hong TS, Wo JY, Roberts H, Beechem JM, Castillo CFD, Mino-Kenudson M, Ting DT, Hemberg M, Hwang WL. Therapy-associated remodeling of pancreatic cancer revealed by single-cell spatial transcriptomics and optimal transport analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.28.546848. [PMID: 37425692 PMCID: PMC10327107 DOI: 10.1101/2023.06.28.546848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
In combination with cell intrinsic properties, interactions in the tumor microenvironment modulate therapeutic response. We leveraged high-plex single-cell spatial transcriptomics to dissect the remodeling of multicellular neighborhoods and cell-cell interactions in human pancreatic cancer associated with specific malignant subtypes and neoadjuvant chemotherapy/radiotherapy. We developed Spatially Constrained Optimal Transport Interaction Analysis (SCOTIA), an optimal transport model with a cost function that includes both spatial distance and ligand-receptor gene expression. Our results uncovered a marked change in ligand-receptor interactions between cancer-associated fibroblasts and malignant cells in response to treatment, which was supported by orthogonal datasets, including an ex vivo tumoroid co-culture system. Overall, this study demonstrates that characterization of the tumor microenvironment using high-plex single-cell spatial transcriptomics allows for identification of molecular interactions that may play a role in the emergence of chemoresistance and establishes a translational spatial biology paradigm that can be broadly applied to other malignancies, diseases, and treatments.
Collapse
Affiliation(s)
- Carina Shiau
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jingyi Cao
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Dennis Gong
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard-MIT Health Sciences and Technology Program, Cambridge, MA, USA
| | - Xunqin Yin
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jae-Won Cho
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter L Wang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jennifer Su
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Steven Wang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | | - Jimmy A Guo
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Biological and Biomedical Sciences Program, Harvard Medical School, Boston, MA, USA
| | - Nicole A Lester
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Jamie L Barth
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tyler Jacks
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Motaz Qadan
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jennifer Y Wo
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hannah Roberts
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David T Ting
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martin Hemberg
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - William L Hwang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
7
|
Yang H, Tan S, Qiao J, Xu Y, Gui Z, Meng Y, Dong B, Peng G, Ibhagui OY, Qian W, Lu J, Li Z, Wang G, Lai J, Yang L, Grossniklaus HE, Yang JJ. Non-invasive detection and complementary diagnosis of liver metastases via chemokine receptor 4 imaging. Cancer Gene Ther 2022; 29:1827-1839. [PMID: 35145271 PMCID: PMC9363530 DOI: 10.1038/s41417-022-00433-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/12/2021] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
Abstract
Noninvasive detection of early-stage liver metastases from different primary cancers is a pressing unmet medical need. The lack of both molecular biomarkers and the sensitive imaging methodology makes the detection challenging. In this study, we observed the elevated expression of chemokine receptor 4 (CXCR4) in uveal melanoma (UM) patient liver tissues, and high CXCR4 expression in liver metastases of UM murine models, regardless of the expression levels in the primary tumors. Based on these findings, we identified CXCR4 as an imaging biomarker and exploited a CXCR4-targeted MRI contrast agent ProCA32.CXCR4 for molecular MRI imaging. ProCA32.CXCR4 has strong CXCR4 binding affinity, high metal selectivity, and r1 and r2 relaxivities, which enables the sensitive detection of liver micrometastases. The MRI imaging capacity for detecting liver metastases was demonstrated in three UM models and one ovarian cancer model. The imaging results were validated by histological and immunohistochemical analysis. ProCA32.CXCR4 has strong potential clinical application for non-invasive diagnosis of liver metastases.
Collapse
Affiliation(s)
- Hua Yang
- Department of Ophthalmology, Emory University, Atlanta, GA, 30322, USA
| | - Shanshan Tan
- Department of Chemistry, Georgia State University, Atlanta, GA, 30303, USA
| | - Jingjuan Qiao
- Department of Chemistry, Georgia State University, Atlanta, GA, 30303, USA
| | - Yiting Xu
- Department of Chemistry, Georgia State University, Atlanta, GA, 30303, USA
| | - Zongxiang Gui
- Department of Chemistry, Georgia State University, Atlanta, GA, 30303, USA
| | - Yuguang Meng
- Yerkes National Primate Research Center, Atlanta, GA, 30329, USA
| | - Bin Dong
- Department of Chemistry, Georgia State University, Atlanta, GA, 30303, USA
| | - Guangda Peng
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
| | | | - Weiping Qian
- Department of Surgery, Emory University, Atlanta, GA, 30322, USA
| | - Jimmy Lu
- Codex BioSolutions Inc, Gaithersburg, MD, USA
| | - Zezhong Li
- Department of Ophthalmology, Emory University, Atlanta, GA, 30322, USA
| | - Guimin Wang
- Department of Ophthalmology, Emory University, Atlanta, GA, 30322, USA
- Affiliated Eye Hospital of Shandong Traditional Chinese Medicine University, Jinan, China
| | - Jinping Lai
- Department of Pathology and Laboratory Medicine, Kaiser Permanente Sacramento Medical Center, Sacramento, CA, 95825, USA
| | - Lily Yang
- Department of Surgery, Emory University, Atlanta, GA, 30322, USA
| | | | - Jenny J Yang
- Department of Chemistry, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
8
|
Current Status of 68Ga-Pentixafor in Solid Tumours. Diagnostics (Basel) 2022; 12:diagnostics12092135. [PMID: 36140541 PMCID: PMC9497673 DOI: 10.3390/diagnostics12092135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/26/2022] [Indexed: 12/24/2022] Open
Abstract
Chemokine receptor CXCR4 is overexpressed in neoplasms and its expression is related to tumour invasion, metastasis and aggressiveness. 68Ga-Pentixafor is used to non-invasively image the expression of CXCR4 in tumours and has been widely used in haematological malignancies. Recent evidence shows that therapies targeting CXCR4 can increase the chemosensitivity of the tumour as well as inhibit tumour metastasis and aggressiveness. 68Ga-Pentixafor has shown promise as an elegant radiotracer to aid in the selection of patients whose tumours demonstrate CXCR4 overexpression and who therefore may benefit from novel therapies targeting CXCR4. In addition, its therapeutic partners 177Lu- and 90Y-Pentixather have been investigated in the treatment of patients with advanced haematological malignancies, and initial studies have shown a good treatment response in metabolically active lesions. 68Ga-Pentixafor in solid tumours complements 18F-FDG by providing prognostic information and selecting patients who may benefit from therapies targeting CXCR4. This review summarises the available literature on the potential applications of 68Ga-Pentixafor in solid tumours.
Collapse
|
9
|
Walther F, Berther JL, Lalos A, Ramser M, Eichelberger S, Mechera R, Soysal S, Muenst S, Posabella A, Güth U, Stadlmann S, Terracciano L, Droeser RA, Zeindler J, Singer G. High ratio of pCXCR4/CXCR4 tumor infiltrating immune cells in primary high grade ovarian cancer is indicative for response to chemotherapy. BMC Cancer 2022; 22:376. [PMID: 35397601 PMCID: PMC8994232 DOI: 10.1186/s12885-022-09374-x] [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: 10/18/2021] [Accepted: 03/04/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Ovarian cancer (OC) is the fifth most common malignant female cancer with a high mortality, mainly because of aggressive high-grade serous carcinomas (HGSOC), but also due to absence of specific early symptoms and effective detection strategies. The CXCL12-CXCR4 axis is considered to have a prognostic impact and to serve as potential therapeutic target. Therefore we investigated the role of pCXCR4 and CXCR4 expression of the tumor cells and of tumor infiltrating immune cells (TIC) in high-grade serous OC and their association with the recurrence-free (RFS) and overall survival (OS).
Methods
A tissue microarray of 47 primary high grade ovarian serous carcinomas and their recurrences was stained with primary antibodies directed against CXCR4 and pCXCR4. Beside the evaluation of the absolute tumor as well as TIC expression in primary and recurrent cancer biopsies the corresponding ratios for pCXCR4 and CXCR4 were generated and analyzed. The clinical endpoints were response to chemotherapy, OS as well as RFS.
Results
Patients with a high pCXCR4/CXCR4 TIC ratio in primary cancer biopsies showed a significant longer RFS during the first two years (p = 0.025). However, this effect was lost in the long-term analysis including a follow-up period of 5 years (p = 0.128). Interestingly, the Multivariate Cox regression analysis showed that a high pCXCR4/CXCR4 TIC ratio in primary cancer independently predicts longer RFS (HR 0.33; 95CI 0.13 - 0.81; p = 0.015). Furthermore a high dichotomized distribution of CXCR4 positive tumor expression in recurrent cancer biopsies showed a significantly longer 6-month RFS rate (p = 0.018) in comparison to patients with low CXCR4 positive tumor expression. However, this effect was not independent of known risk factors in a Multivariate Cox regression (HR 0.57; 95CI 0.24 - 1.33; p = 0.193).
Conclusions
To the best of our knowledge we show for the first time that a high pCXCR4/CXCR4 TIC ratio in primary HGSOC biopsies is indicative for better RFS and response to chemotherapy.
Highlights
• We observed a significant association between high pCXCR4/CXCR4 TIC ratio and better RFS in primary cancer biopsies, especially during the early postoperative follow-up and independent of known risk factors for recurrence.
• High CXCR4 tumor expression in recurrent HGSOC biopsies might be indicative for sensitivity to chemotherapy. We found evidence that at the beginning of the disease (early follow-up) the role of the immune response seems to be the most crucial factor for progression. On the other hand in recurrent/progressive disease the biology of the tumor itself becomes more important for prognosis.
• We explored for the first time the predictive and prognostic role of pCXCR4/CXCR4 TIC ratio in high-grade serous ovarian cancer.
Collapse
|
10
|
CXC Chemokine Signaling in Progression of Epithelial Ovarian Cancer: Theranostic Perspectives. Int J Mol Sci 2022; 23:ijms23052642. [PMID: 35269786 PMCID: PMC8910147 DOI: 10.3390/ijms23052642] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 02/06/2023] Open
Abstract
Patients with epithelial ovarian cancer (EOC) are often diagnosed at an advanced stage due to nonspecific symptoms and ineffective screening approaches. Although chemotherapy has been available and widely used for the treatment of advanced EOC, the overall prognosis remains dismal. As part of the intrinsic defense mechanisms against cancer development and progression, immune cells are recruited into the tumor microenvironment (TME), and this process is directed by the interactions between different chemokines and their receptors. In this review, the functional significance of CXC chemokine ligands/chemokine receptors (CXCL/CXCR) and their roles in modulating EOC progression are summarized. The status and prospects of CXCR/CXCL-based theranostic strategies in EOC management are also discussed.
Collapse
|
11
|
Liu X, Ma B, Chen M, Zhang Y, Ma Z, Chen H. Prognostic Autophagy-Related Genes of Gastric Cancer Patients on Chemotherapy. Front Genet 2021; 12:720849. [PMID: 34759953 PMCID: PMC8573096 DOI: 10.3389/fgene.2021.720849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Chemotherapy resistance based on fluorouracil and cisplatin is one of the most encountered postoperative clinical problems in patients diagnosed with gastric cancer (GC), resulting in poor prognosis. Aim of the Study: This study aimed to combine autophagy-related genes (ARGs) to investigate the susceptibility patients with GC to postoperative chemotherapy. Methods: Based on The Cancer Genome Atlas (TCGA) database, gene expression data for GC patients undergoing chemotherapy were integrated and analyzed. Prognostic genes were screened based on univariate and multivariate analysis regression analysis. Subjects were divided into high-risk and low-risk groups according to the median risk score. Kaplan-Meier method was used to evaluate OS and DFS. The accuracy of the prediction was determined by the subject operating characteristic curve analysis. In addition, stratified analyses based on different clinical variables was performed to assess the correlation between risk scores and clinical variables. Quantitative real-time (qRT) PCR was used to verify the expression of CXCR4 in GC tissues and cell lines. Results: A total of nine ARGs related to the prognosis of chemotherapy patients were screened out. Compared with normal gastric mucosa cell, CXCR4 showed elevated expression in GC and was significantly associated with survival. Based on GEO and TCGA databases, the model accurately predicted DFS and OS after chemotherapy. Conclusion: This study established prognostic markers based on nine genes, predicting that ARGs are related to chemotherapy susceptibility of GC patients, which can provide better individualized treatment regimens for clinical practice.
Collapse
Affiliation(s)
- Xiaolong Liu
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China.,Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Bin Ma
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Mali Chen
- Department of Obstetrics, Gansu Province Maternity and Child-Care Hospital, Lanzhou, China
| | - Yaqing Zhang
- Department of Gynaecology, Gansu Province Maternity and Child-Care Hospital, Lanzhou, China
| | - Zhen Ma
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China.,Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Hao Chen
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China.,Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
12
|
Gamal AA, Abbas HY, Abdelwahed NAM, Kashef MT, Mahmoud K, Esawy MA, Ramadan MA. Optimization strategy of Bacillus subtilis MT453867 levansucrase and evaluation of levan role in pancreatic cancer treatment. Int J Biol Macromol 2021; 182:1590-1601. [PMID: 34015407 DOI: 10.1016/j.ijbiomac.2021.05.056] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
Pancreatic cancer is the fourth most lethal cancer type worldwide. Due to multiple levan applications including anticancer activities, studies related to levansucrase production are of interest. To our knowledge, levan effect on pancreatic cancer cells has not been tested previously. In this work, among eighteen bacterial honey isolates, Bacillus subtilis MT453867 showed the highest levan yield (33 g/L) and levansucrase production (8.31 U/mL). One-factor-at-a-time technique increased levansucrase activity by 60% when MgSO4 was eliminated. The addition of 60 g/L banana peels enhanced the enzyme activity (192 U/mL). Placket Burman design determined the media composition for maximum levan yield (54.8 g/L) and levansucrase production (505 U/mL). The identification of levan was confirmed by thin-layer chromatography, Fourier-Transform Infrared spectrometric analysis, 13C-nuclear-magnetic resonance, and 1H-nuclear-magnetic resonance. Both crude and dialyzed levan completely inhibited the pancreatic cancer cell line at 100 ppm with no cytotoxicity on the normal retinal cell line. The LD50 of crude levan was 4833 mg/kg body weight. Levan had strong antioxidant activity and significantly reduced the expression of CXCR4 and MCM7 genes in pancreatic cancer cells with significant DNA fragmentation. In conclusion, Bacillus subtilis MT453867 levan is a promising adjunct to pancreatic-anticancer agents with both anti-cancer and chemoprotective effects.
Collapse
Affiliation(s)
- Amira A Gamal
- Chemistry of Natural and Microbial Products Department, Pharmaceutical Industries Research Division, National Research Centre, 33 El Bohouth st.(former El Tahrir st.), P.O 12622, Dokki, Cairo, Egypt
| | - Heba Y Abbas
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Sadat City
| | - Nayera A M Abdelwahed
- Chemistry of Natural and Microbial Products Department, Pharmaceutical Industries Research Division, National Research Centre, 33 El Bohouth st.(former El Tahrir st.), P.O 12622, Dokki, Cairo, Egypt
| | - Mona T Kashef
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University
| | - Khaled Mahmoud
- Pharmacognosy Department, Pharmaceutical Industries Research Division, National Research Centre, 33 El Bohouth st.(former El Tahrir st.), P.O 12622, Dokki, Cairo, Egypt
| | - Mona A Esawy
- Chemistry of Natural and Microbial Products Department, Pharmaceutical Industries Research Division, National Research Centre, 33 El Bohouth st.(former El Tahrir st.), P.O 12622, Dokki, Cairo, Egypt.
| | - Mohammed A Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University
| |
Collapse
|
13
|
Linde P, Baues C, Wegen S, Trommer M, Quaas A, Rosenbrock J, Celik E, Marnitz S, Bruns CJ, Fischer T, Schomaecker K, Wester HJ, Drzezga A, van Heek L, Kobe C. Pentixafor PET/CT for imaging of chemokine receptor 4 expression in esophageal cancer - a first clinical approach. Cancer Imaging 2021; 21:22. [PMID: 33579381 PMCID: PMC7881561 DOI: 10.1186/s40644-021-00391-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/05/2021] [Indexed: 12/24/2022] Open
Abstract
Background Expression of CXCR4, a chemokine (C-X-C motif) receptor that plays a central role in tumor growth and metastasis of circulating tumor cells, has been described in a variety of solid tumors. A high expression of CXCR4 has a prognostic significance with regard to overall and progression-free survival and offers a starting point for targeted therapies. In this context, [68]Ga-Pentixafor-Positron Emission Tomography/Computer Tomography (PET/CT) offers promising possibility of imaging the CXCR4 expression profile. We set out to compare a [18F] fluorodeoxyglucose (FDG)-PET/CT and a [68Ga]Pentixafor-PET/CT in (re-)staging and radiation planning of patients with localized esophageal cancer. Materials and methods In this retrospective analysis, ten patients, with adeno- or squamous cell carcinoma of the esophagus (n = 3 and n = 7, respectively), which were scheduled for radio (chemo) therapy, were imaged using both Pentixafor and FDG PET/CT examinations. All lesions were visually rated as Pentixafor and FDG positive or negative. For both tracers, SUVmax was measured all lesions and compared to background. Additionally, immunohistochemistry of CXCR4 was obtained in patients undergoing surgery. Results FDG-positive tumor-suspicious lesions were detected in all patients and a total of 26 lesions were counted. The lesion-based analysis brought equal status in 14 lesions which were positive for both tracers while five lesions were FDG positive and Pentixafor negative and seven lesions were FDG negative, but Pentixafor positive. Histopathologic correlation was available in seven patients. The CXCR4 expression of four non-pretreated tumour lesion samples was confirmed immunohistochemically. Conclusion Our data shows that additional PET/CT imaging with Pentixafor for imaging the CXCR4 chemokine receptor is feasible but heterogeneous in both newly diagnosed and pretreated recurrent esophageal cancer. In addition, the Pentixafor PET/CT may serve as complementary tool for radiation field expansion in radiooncology. Supplementary Information The online version contains supplementary material available at 10.1186/s40644-021-00391-w.
Collapse
Affiliation(s)
- Philipp Linde
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany.
| | - Christian Baues
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Simone Wegen
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Maike Trommer
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Alexander Quaas
- Department of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Johannes Rosenbrock
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Eren Celik
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Simone Marnitz
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Christiane J Bruns
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Thomas Fischer
- Department of Nuclear Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Klaus Schomaecker
- Department of Nuclear Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Hans-Juergen Wester
- Department of Radiochemistry, Technische Universität München, Garching, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Lutz van Heek
- Department of Nuclear Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Carsten Kobe
- Department of Nuclear Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
14
|
Sad LAE, Mohamed D, Elanwar N, Elkady A. CXCR4 and RIF1 overexpression induces resistance of epithelial ovarian cancer to cisplatin-based chemotherapy. J Cancer Res Ther 2021; 17:1454-1461. [DOI: 10.4103/jcrt.jcrt_480_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
15
|
Hao Y, Miao J, Liu W, Peng L, Chen Y, Zhong Q. Formononetin protects against cisplatin‑induced acute kidney injury through activation of the PPARα/Nrf2/HO‑1/NQO1 pathway. Int J Mol Med 2020; 47:511-522. [PMID: 33416097 PMCID: PMC7797437 DOI: 10.3892/ijmm.2020.4805] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/22/2020] [Indexed: 12/15/2022] Open
Abstract
Acute kidney injury (AKI) is characterized by an abrupt deterioration of renal function. Formononetin (FOR) protects against cisplatin (CIS)‑induced AKI, and it has various potential pharmacological and biological effects, including anti‑inflammatory, antioxidative and anti‑apoptotic effects. The current study investigated the role of FOR in CIS‑induced AKI. Rats were treated with CIS to establish an AKI model, followed by treatment with FOR. HK‑2 cells were treated with CIS, FOR, GW6471 [a peroxisome proliferator‑activated receptor α (PPARα) antagonist], eupatilin (a PPARα agonist) and nuclear factor erythroid 2‑related factor 2 (Nrf2) small interfering RNA (siNrf2), and cell proliferation and apoptosis were determined by MTT and flow cytometry assays. The mRNA and proteins levels of PPARα, Nrf2, heme oxygenase‑1 (HO‑1) and NAD(P)H quinone dehydrogenase 1 (NQO1) were measured by reverse transcription‑quantitative PCR and western blotting. The results demonstrated that FOR attenuated the histopathological changes, the levels of blood urea nitrogen, creatinine, TNF‑α and IL‑1β, and the MDA content and MPO activity, whereas it enhanced CAT activity in the AKI rat model. Furthermore, FOR and eupatilin promoted cell viability and CAT activity, and increased the levels of PPARα, Nrf2 and HO‑1 and NQO1, but suppressed apoptosis and MPO activity, and reduced the levels of MDA, TNF‑α and IL‑1β in CIS‑treated HK‑2 cells. Notably, the aforementioned effects were reversed by GW6471 treatment or siNrf2 transfection. In conclusion, FOR protects against CIS‑induced AKI via activation of the PPARα/Nrf2/HO‑1/NQO1 pathway.
Collapse
Affiliation(s)
- Yan Hao
- Department of Nephrology, Zigong First People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Jie Miao
- Department of Imaging Medicine, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan 643000, P.R. China
| | - Wenjia Liu
- Department of Nephrology, Zigong First People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Li Peng
- Department of Nephrology, Zigong First People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yue Chen
- Department of Nephrology, Zigong First People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Qing Zhong
- Department of Nephrology, Zigong First People's Hospital, Zigong, Sichuan 643000, P.R. China
| |
Collapse
|
16
|
Liu J, Meng H, Nie S, Sun Y, Jiang P, Li S, Yang J, Sun R, Cheng W. Identification of a prognostic signature of epithelial ovarian cancer based on tumor immune microenvironment exploration. Genomics 2020; 112:4827-4841. [PMID: 32890701 DOI: 10.1016/j.ygeno.2020.08.027] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/24/2020] [Accepted: 08/21/2020] [Indexed: 12/22/2022]
Abstract
This study aims to develop an immune-related genes (IRGs) prognostic signature to stratify the epithelial ovarian cancer (EOC) patients. We identified 332 up- and 154 down-regulated EOC-specific IRGs. As a result, candidate IRGs were idendified to construct prognostic models respectivy for overall survial and progression-free survival. The risk score was validated as a risk factor for prognosis and was used to built a combined nomogram. According to the IRG-related prognostic model, EOC patients were divided into high- and low- risk group and were further explored their association with tumor immune microenvironment (TME). CIBERSORT algorithm showed higher macrophages M1 cell, T cells follicular helper cell and plasma cells infiltrating levels in the low-risk group. In addition, the low-risk group was found with higher immunophenoscore and distinct mutation signatures compared with the high-risk group. These findings may shed light on the development of novel immune biomarkers and target therapy of EOC.
Collapse
Affiliation(s)
- Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Huangyang Meng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Sipei Nie
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Ying Sun
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Pinping Jiang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Siyue Li
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Jing Yang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Rui Sun
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Wenjun Cheng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
17
|
Behera A, Ashraf R, Srivastava AK, Kumar S. Bioinformatics analysis and verification of molecular targets in ovarian cancer stem-like cells. Heliyon 2020; 6:e04820. [PMID: 32984578 PMCID: PMC7492822 DOI: 10.1016/j.heliyon.2020.e04820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/01/2020] [Accepted: 08/26/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is a lethal and aggressive gynecological malignancy. Despite recent advances, existing therapies are challenged by a high relapse rate, eventually resulting in disease recurrence and chemoresistance. Emerging evidence indicates that a subpopulation of cells known as cancer stem-like cells (CSLCs) exists with non-tumorigenic cancer cells (non-CSCs) within a bulk tumor and is thought to be responsible for tumor recurrence and drug-resistance. Therefore, identifying the molecular drivers for cancer stem cells (CSCs) is critical for the development of novel therapeutic strategies for the treatment of EOC. METHODS Two gene datasets were downloaded from the Gene Expression Omnibus (GEO) database based on our search criteria. Differentially expressed genes (DEGs) in both datasets were obtained by the GEO2R web tool. Based on log2 (fold change) >2, the top thirteen up-regulated genes and log2 (fold change) < -1.5 top thirteen down-regulated genes were selected, and the association between their expressions and overall survival was analyzed by OncoLnc web tool. Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome pathways analysis, and protein-protein interaction (PPI) networks were performed for all the common DEGs found in both datasets. SK-OV-3 cells were cultured in an adherent culture medium and spheroids were generated in suspension culture with CSCs specific medium. RNA from both cell population was extracted to validate the selected DEGs expression by q-PCR. Growth inhibition assay was performed in SK-OV-3 cells after carboplatin treatment. RESULTS A total of 200 DEGs, 117 up-regulated and 83 down-regulated genes were commonly identified in both datasets. Analysis of pathways and enrichment tests indicated that the extracellular matrix part, cell proliferation, tissue development, and molecular function regulation were enriched in CSCs. Biological pathways such as interferon-alpha/beta signaling, molecules associated with elastic fibers, and synthesis of bile acids and bile salts were significantly enriched in CSCs. Among the top 13 up-regulated and down-regulated genes, MMP1 and PPFIBP1 expression were associated with overall survival. Higher expression of ADM, CXCR4, LGR5, and PTGS2 in carboplatin treated SK-OV-3 cells indicate a potential role in drug resistance. CONCLUSIONS The molecular signature and signaling pathways enriched in ovarian CSCs were identified by bioinformatics analysis. This analysis could provide further research ideas to find the new mechanism and novel potential therapeutic targets for ovarian CSCs.
Collapse
Affiliation(s)
- Abhijeet Behera
- Division of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, India
| | - Rahail Ashraf
- Division of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, India
| | - Amit Kumar Srivastava
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India
| | - Sanjay Kumar
- Division of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, India
| |
Collapse
|
18
|
Zhang K, Zhou H, Yan B, Cao X. TUG1/miR-133b/CXCR4 axis regulates cisplatin resistance in human tongue squamous cell carcinoma. Cancer Cell Int 2020; 20:148. [PMID: 32390763 PMCID: PMC7201732 DOI: 10.1186/s12935-020-01224-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023] Open
Abstract
Background Long noncoding RNA taurine upregulated 1 (TUG1) has been reported to play an important role in human cancers. However, little is known about the role of TUG1 in drug resistance and its mechanism in tongue squamous cell carcinoma (TSCC). Methods Twenty-one cisplatin-sensitive or resistant TSCC patients were enrolled in this study. Cisplatin-resistant cells (SCC25/CDDP and CAL27/CDDP) were used for experiments in vitro. Transfection was performed using Lipofectamine 2000 transfection reagent. The levels of TUG1, microRNA-133b (miR-133b) and cysteine-X-cysteine chemokine receptor 4 (CXCR4) were measured by quantitative real-time polymerase chain reaction or western blot. The cisplatin resistance was investigated by cell viability, transwell invasion and apoptosis assays. The interactions among TUG1, miR-133b and CXCR4 were evaluated by luciferase reporter assay and RNA immunoprecipitation. Murine xenograft model was established using the stably transfected CAL27/CDDP cells. Results TUG1 expression was elevated in cisplatin-resistant TSCC tissues and cells compared with that in sensitive group and its knockdown inhibited cisplatin resistance to SCC25/CDDP and CAL27/CDDP cells. miR-133b was targeted via TUG1 and its overexpression suppressed cisplatin resistance. Moreover, CXCR4 was a target of miR-133b. CXCR4 silence repressed cisplatin resistance, which was reversed by miR-133b knockdown. The level of CXCR4 protein was decreased by inhibition of TUG1 and recuperated by miR-133b knockdown. Besides, interference of TUG1 attenuated tumor growth by regulating miR-133b and CXCR4 in vivo. Conclusion Downregulation of TUG1 impeded cisplatin resistance in TSCC-resistant cells by mediating miR-133b and CXCR4, indicating TUG1 as a promising target for TSCC chemotherapy.
Collapse
Affiliation(s)
- Ke Zhang
- The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450000 Henan China
| | - Hong Zhou
- The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450000 Henan China
| | - Bo Yan
- The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450000 Henan China
| | - Xuanping Cao
- The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450000 Henan China
| |
Collapse
|
19
|
Zhang J, Quan LN, Meng Q, Wang HY, Wang J, Yu P, Fu JT, Li YJ, Chen J, Cheng H, Wu QP, Yu XR, Yun HY, Huang SG. miR-548e Sponged by ZFAS1 Regulates Metastasis and Cisplatin Resistance of OC by Targeting CXCR4 and let-7a/BCL-XL/S Signaling Axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 20:621-638. [PMID: 32353736 PMCID: PMC7191130 DOI: 10.1016/j.omtn.2020.03.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/17/2020] [Accepted: 03/27/2020] [Indexed: 12/23/2022]
Abstract
Ovarian cancer (OC) is a severe malignancy featuring a poor prognosis due to rapid metastasis and chemotherapy resistance. In this study, we extensively investigated the upstream and downstream mechanisms of miR-548e in regulating OC progression and cisplatin resistance. Our results indicated that ZFAS1 was highly expressed and promoted OC cell proliferation, migration, invasion, and cisplatin resistance by directly suppressing miR-548e expression. ZFAS1 co-localized with miR-548e in the cytosols of OC cells. miR-548e repressed CXCR4 expression, and elevated CXCR4 expression promoted OC cell proliferation, migration, invasion, and cisplatin resistance. Cisplatin resistance induced by ZFAS1 and CXCR4 overexpression in OC cells was mediated by their suppression on let-7a and elevation of BCL-XL/S expression. ZFAS1 knockdown and miR-548e and let-7a overexpression impaired cisplatin resistance and suppressed lung metastatic nodule formation in nude mice. In conclusion, ZFAS1 binds with miR-548e to enhance CXCR4 expression to promote OC cell proliferation and metastasis, which also enhances cisplatin resistance by suppressing let-7a and elevating BCL-XL/S protein expression.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Li-Ni Quan
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Qiu Meng
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Hai-Yan Wang
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Jie Wang
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Pin Yu
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Jian-Tao Fu
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Ying-Jia Li
- Clinical Laboratory, Third Xiangya Hospital of Central South University, No. 138 Tong Zipo Road, Changsha 410013, Hunan Province, P.R. China
| | - Jin Chen
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Hong Cheng
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Qing-Ping Wu
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Xin-Rong Yu
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Hong-Ye Yun
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China
| | - Shou-Guo Huang
- Department of Obstetrics and Gynecology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Road, Haidian Island, Haikou 570208, Hainan Province, P.R. China.
| |
Collapse
|
20
|
Therapy-Induced Changes in CXCR4 Expression in Tumor Xenografts Can Be Monitored Noninvasively with N-[ 11C]Methyl-AMD3465 PET. Mol Imaging Biol 2019; 22:883-890. [PMID: 31802362 PMCID: PMC7343732 DOI: 10.1007/s11307-019-01447-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Purpose Chemokine CXCL12 and its receptor CXCR4 are constitutively overexpressed in human cancers. The CXCL12-CXCR4 signaling axis plays an important role in tumor progression and metastasis, but also in treatment-induced recruitment of CXCR4-expressing cytotoxic immune cells. Here, we aimed to demonstrate the feasibility of N-[11C]methyl-AMD3465 positron emission tomography (PET) to monitor changes in CXCR4 density in tumors after single-fraction local radiotherapy or in combination with immunization. Procedure TC-1 cells expressing human papillomavirus antigens E6 and E7 were inoculated into the C57BL/6 mice subcutaneously. Two weeks after tumor cell inoculation, mice were irradiated with a single-fraction 14-Gy dose of X-ray. One group of irradiated mice was immunized with an alpha-viral vector vaccine, SFVeE6,7, and another group received daily injections of the CXCR4 antagonist AMD3100 (3 mg/kg -intraperitoneal (i.p.)). Seven days after irradiation, all animals underwent N-[11C]methyl-AMD3465 PET. Results PET imaging showed N-[11C]methyl-AMD3465 uptake in the tumor of single-fraction irradiated mice was nearly 2.5-fold higher than in sham-irradiated tumors (1.07 ± 0.31 %ID/g vs. 0.42 ± 0.05 % ID/g, p < 0.01). The tumor uptake was further increased by 4-fold (1.73 ± 0.17 % ID/g vs 0.42 ± 0.05 % ID/g, p < 0.01) in mice treated with single-fraction radiotherapy in combination with SFVeE6,7 immunization. Administration of AMD3100 caused a 4.5-fold reduction in the tracer uptake in the tumor of irradiated animals (0.24 ± 0.1 % ID/g, p < 0.001), suggesting that tracer uptake is indeed due to CXCR4-mediated chemotaxis. Conclusion This study demonstrates the feasibility of N-[11C]methyl-AMD3465 PET imaging to monitor treatment-induced changes in the density of CXCR4 receptors in tumors and justifies further evaluation of CXCR4 as a potential imaging biomarker for evaluation of anti-tumor therapies.
Collapse
|
21
|
Lee NH, Nikfarjam M, He H. Functions of the CXC ligand family in the pancreatic tumor microenvironment. Pancreatology 2018; 18:705-716. [PMID: 30078614 DOI: 10.1016/j.pan.2018.07.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/06/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023]
Abstract
Therapeutic resistance is the major contributor to the poor prognosis of and low survival from pancreatic cancer (PC). Cancer progression is a complex process reliant on interactions between the tumor and the tumor microenvironment (TME). Members of the CXCL family of chemokines are present in the pancreatic TME and seem to play a vital role in regulating PC progression. As pancreatic tumors interact with the TME and with PC stem cells (CSCs), determining the roles of specific members of the CXCL family is vital to the development of improved therapies. This review highlights the roles of selected CXCLs in the interactions between pancreatic tumor and its stroma, and in CSC phenotypes, which can be used to identify potential treatment targets.
Collapse
Affiliation(s)
- Nien-Hung Lee
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Mehrdad Nikfarjam
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Hong He
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia.
| |
Collapse
|
22
|
Hudson LG, Gillette JM, Kang H, Rivera MR, Wandinger-Ness A. Ovarian Tumor Microenvironment Signaling: Convergence on the Rac1 GTPase. Cancers (Basel) 2018; 10:cancers10100358. [PMID: 30261690 PMCID: PMC6211091 DOI: 10.3390/cancers10100358] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment for epithelial ovarian cancer is complex and rich in bioactive molecules that modulate cell-cell interactions and stimulate numerous signal transduction cascades. These signals ultimately modulate all aspects of tumor behavior including progression, metastasis and therapeutic response. Many of the signaling pathways converge on the small GTPase Ras-related C3 botulinum toxin substrate (Rac)1. In addition to regulating actin cytoskeleton remodeling necessary for tumor cell adhesion, migration and invasion, Rac1 through its downstream effectors, regulates cancer cell survival, tumor angiogenesis, phenotypic plasticity, quiescence, and resistance to therapeutics. In this review we discuss evidence for Rac1 activation within the ovarian tumor microenvironment, mechanisms of Rac1 dysregulation as they apply to ovarian cancer, and the potential benefits of targeting aberrant Rac1 activity in this disease. The potential for Rac1 contribution to extraperitoneal dissemination of ovarian cancer is addressed.
Collapse
Affiliation(s)
- Laurie G Hudson
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Jennifer M Gillette
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Huining Kang
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Melanie R Rivera
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Angela Wandinger-Ness
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
23
|
Fong P, Ao CN, Tou KI, Huang KM, Cheong CC, Meng LR. Experimental and In Silico Analysis of Cordycepin and its Derivatives as Endometrial Cancer Treatment. Oncol Res 2018; 27:237-251. [PMID: 29673423 PMCID: PMC7848235 DOI: 10.3727/096504018x15235274183790] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The aim of this study was to investigate the inhibition effects of cordycepin and its derivatives on endometrial cancer cell growth. Cytotoxicity MTT assays, clonogenic assays, and flow cytometry were used to observe the effects on apoptosis and regulation of the cell cycle of Ishikawa cells under various concentrations of cordycepin, cisplatin, and combinations of the two. Validated in silico docking simulations were performed on 31 cordycepin derivatives against adenosine deaminase (ADA) to predict their binding affinities and hence their potential tendency to be metabolized by ADA. Cordycepin has a significant dose-dependent inhibitory effect on cell proliferation. The combination of cordycepin and cisplatin produced greater inhibition effects than did cordycepin alone. Apoptosis investigations confirmed the ability of cordycepin to induce the apoptosis of Ishikawa cells. The in silico results indicate that compound MRS5698 is least metabolized by ADA and has acceptable drug likeness and safety profiles. This is the first study to confirm the cytotoxic effects of cordycepin on endometrial cancer cells. This study also identified cordycepin derivatives with promising pharmacological and pharmacokinetic properties for further investigation in the development of new treatments for endometrial cancer.
Collapse
Affiliation(s)
- Pedro Fong
- School of Health Sciences, Macao Polytechnic Institute, Macao, P.R. China
| | - Cheng N Ao
- School of Health Sciences, Macao Polytechnic Institute, Macao, P.R. China
| | - Kai I Tou
- School of Health Sciences, Macao Polytechnic Institute, Macao, P.R. China
| | - Ka M Huang
- School of Health Sciences, Macao Polytechnic Institute, Macao, P.R. China
| | - Chi C Cheong
- School of Health Sciences, Macao Polytechnic Institute, Macao, P.R. China
| | - Li R Meng
- School of Health Sciences, Macao Polytechnic Institute, Macao, P.R. China
| |
Collapse
|
24
|
Li Q, Zhang J, Zhou J, Yang B, Liu P, Cao L, Jing L, Liu H. lncRNAs are novel biomarkers for differentiating between cisplatin-resistant and cisplatin-sensitive ovarian cancer. Oncol Lett 2018; 15:8363-8370. [PMID: 29805570 PMCID: PMC5950027 DOI: 10.3892/ol.2018.8433] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 12/05/2017] [Indexed: 12/16/2022] Open
Abstract
Cisplatin-resistant ovarian cancer occurs in patients with ovarian cancer treated with cisplatin-based chemotherapy, which results in tumor progression during treatment, or recurrence of the tumor within 6 months of the treatment. It is vital that a novel biomarker for diagnosis, or an efficient therapeutic target of cisplatin-resistant ovarian is identified. Long non-coding (lnc)RNAs were determined to serve critical functions in a variety of distinct types of cancer, including ovarian cancer; however, there is limited knowledge regarding the differential expression levels of lncRNAs in cisplatin-resistant and cisplatin-sensitive ovarian cancer. Therefore, in the present study, the expression levels were determined for these cancer types. The lncRNA expression profile in cisplatin-resistant ovarian cancer was analyzed and compared with the results for cisplatin-sensitive ovarian cancer; gene ontology and pathway analysis demonstrated that the dysregulated lncRNAs participated in important biological processes. Subsequently, it was identified that these dysregulated lncRNAs were present in other ovarian cancer tissues and in SKOV3 ovarian cancer cells, as well as its cisplatin-resistant clone, SKOV3/CDDP. In addition, it was revealed that 8 lncRNAs (Enst0000435726, Enst00000585612, Enst00000566734, Enst00000453783, NR_023915, RP11_697E22.2, uc010jub.1 and tcons_00008505) were associated with cisplatin-resistant ovarian cancer. The present study may assist in improving understanding of the initiation and developmental mechanisms underlying cisplatin-resistant ovarian cancer, which could aid future studies in discovering potential biomarkers for diagnosis or therapeutic targets that may be used in clinical treatment.
Collapse
Affiliation(s)
- Qing Li
- Department of Pathology, Shanghai Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| | - Juan Zhang
- Department of Pathology, Affiliated Nanjing Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Juan Zhou
- Department of Pathology, Shanghai Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| | - Binglie Yang
- Department of Gynecology and Obstetrics, Shanghai Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| | - Pingping Liu
- Department of Gynecology and Obstetrics, Shanghai Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| | - Lei Cao
- Department of Pathology, Shanghai Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| | - Lei Jing
- Department of Pathology, Shanghai Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| | - Hua Liu
- Department of Pathology, Shanghai Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| |
Collapse
|
25
|
Figueras A, Alsina-Sanchís E, Lahiguera Á, Abreu M, Muinelo-Romay L, Moreno-Bueno G, Casanovas O, Graupera M, Matias-Guiu X, Vidal A, Villanueva A, Viñals F. A Role for CXCR4 in Peritoneal and Hematogenous Ovarian Cancer Dissemination. Mol Cancer Ther 2017; 17:532-543. [PMID: 29146630 DOI: 10.1158/1535-7163.mct-17-0643] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/08/2017] [Accepted: 11/03/2017] [Indexed: 11/16/2022]
Abstract
Epithelial ovarian cancer is characterized by a low recovery rate because the disease is typically diagnosed at an advanced stage, by which time most patients (80%) already exhibit disseminated neoplasia. The cytokine receptor CXCR4 has been implicated in the development of metastasis in various tumor types. Using a patient-derived tissue macroarray and mRNA expression analysis, we observed high CXCR4 levels in high-grade serous epithelial ovarian carcinomas, the most metastatic tumor, compared with those in endometrioid carcinomas. CXCR4 inhibition by treatment with the CXCR4 antagonist AMD3100 or by expression of shRNA anti-CXCR4 similarly inhibited angiogenesis in several models of ovarian carcinomas orthotopically grown in nude mice, but the effect on tumor growth was correlated with the levels of CXCR4 expression. Moreover, CXCR4 inhibition completely blocked dissemination and metastasis. This effect was associated with reduced levels of active Src, active ERKs, the inhibition of EMT transition, and block of hematogenous ovarian cancer dissemination decreasing circulating human tumoral cells (CTC). In tumors, CXCR4-expressing cells also had more mesenchymal characteristics. In conclusion, our results indicate that CXCR4 expression confers a proinvasive phenotype to ovarian carcinoma cells. Thus, anti-CXCR4 therapy is a possible agent for a complementary treatment of advanced disseminated epithelial high-grade serous ovarian cancer patients. Mol Cancer Ther; 17(2); 532-43. ©2017 AACR.
Collapse
Affiliation(s)
- Agnès Figueras
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Elisenda Alsina-Sanchís
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Álvaro Lahiguera
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Manuel Abreu
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Gema Moreno-Bueno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPaz, Madrid, Spain.,MD Anderson International Foundation, Madrid, Spain
| | - Oriol Casanovas
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Mariona Graupera
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain.,Laboratori d'Oncologia Molecular, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Xavier Matias-Guiu
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Servei d'Anatomia Patològica, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - August Vidal
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain.,Servei d'Anatomia Patològica, Hospital Universitari de Bellvitge, Barcelona, Spain.,Xenopat, Carrer de la Feixa Llarga S/N, L'Hospitalet de Llobregat, Barcelona, Spain.,Departament de Patologia i Terapèutica Experimental, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Alberto Villanueva
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain.,Xenopat, Carrer de la Feixa Llarga S/N, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Francesc Viñals
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain. .,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain.,Departament de Ciències Fisiològiques, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Lee KC, Lin CT, Chang SF, Chen CN, Liu JL, Huang WS. Effect of AICAR and 5-Fluorouracil on X-ray Repair, Cross-Complementing Group 1 Expression, and Consequent Cytotoxicity Regulation in Human HCT-116 Colorectal Cancer Cells. Int J Mol Sci 2017; 18:ijms18112363. [PMID: 29117108 PMCID: PMC5713332 DOI: 10.3390/ijms18112363] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer mortality and 5-Fluorouracil (5-FU) is the most common chemotherapy agent of CRC. A high level of X-ray repair cross complementing group 1 (XRCC1) in cancer cells has been associated with the drug resistance occurrence. Moreover, the activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) has been indicated to regulate the cancer cell survival. Thus, this study was aimed to examine whether XRCC1 plays a role in the 5-FU/AMPK agonist (AICAR)-induced cytotoxic effect on CRC and the underlying mechanisms. Human HCT-116 colorectal cells were used in this study. It was shown that 5-FU increases the XRCC1 expression in HCT-116 cells and then affects the cell survival through CXCR4/Akt signaling. Moreover, 5-FU combined with AICAR further result in more survival inhibition in HCT-116 cells, accompanied with reduced CXCR4/Akt signaling activity and XRCC1 expression. These results elucidate the role and mechanism of XRCC1 in the drug resistance of HCT-116 cells to 5-FU. We also demonstrate the synergistic inhibitory effect of AMPK on 5-FU-inhibited HCT-116 cell survival under the 5-FU and AICAR co-treatment. Thus, our findings may provide a new notion for the future drug regimen incorporating 5-FU and AMPK agonists for the CRC treatment.
Collapse
Affiliation(s)
- Ko-Chao Lee
- Department of Colorectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Kaohsiung Medical Center, Kaohsiung 833, Taiwan.
| | - Chien-Tsong Lin
- Center for General Education, National Formosa University, Yunlin 632, Taiwan.
- Department of Wood Based Materials and Design, National Chiayi University, Chiayi 600, Taiwan.
| | - Shun-Fu Chang
- Department of Medical Research and Development, Chang Gung Memorial Hospital Chiayi Branch, Chiayi 613, Taiwan.
| | - Cheng-Nan Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600, Taiwan.
| | - Jing-Lan Liu
- Department of Pathology, Chang Gung Memorial Hospital Chiayi Branch, Chiayi 600, Taiwan.
| | - Wen-Shih Huang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan.
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi 613, Taiwan.
| |
Collapse
|
27
|
Jain A, Jahagirdar D, Nilendu P, Sharma NK. Molecular approaches to potentiate cisplatin responsiveness in carcinoma therapeutics. Expert Rev Anticancer Ther 2017; 17:815-825. [PMID: 28705091 DOI: 10.1080/14737140.2017.1356231] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cisplatin has been considered as the crucial regimen of widely prescribed chemotherapy treatment for cancer. The advancing treatment of cancers has reached the border line, where tumors show resistance to cisplatin and may thwart its use. Other than issues of drug resistance, cisplatin has been reported to evince side effects such as nephrotoxicity and ototoxicity. Therefore, there is a compelling need to untangle the problems associated with cisplatin treatment in carcinoma. Areas covered: In this review, we summarize the current status of combinatorial options to bring about better pre-clinical and clinical cisplatin drug responses in carcinoma. We begin with problems associated with cisplatin drugs and current avenues such as depicting molecular modulation of enhanced influx and reduced efflux. We also discuss the scope of the DNA damage response landscape and contribution of regulatory small RNAs towards potentiation of cisplatin responses. Expert commentary: The extensive use of cisplatin and incessant high drug dose have prompted the scientific community to limit the burden of cisplatin without compromising therapeutic success. Currently, there are reports on the potential use of other non-toxic small molecule inhibitors, interference RNAs and peptide mimetics to get rid of cellular adversities responsible for cisplatin resistance and high dose effects.
Collapse
Affiliation(s)
- Aayushi Jain
- a Cancer and Translational Research Lab , Dr. D.Y. Patil Biotechnology & Bioinformatics Institute , Pune , India
| | - Devashree Jahagirdar
- a Cancer and Translational Research Lab , Dr. D.Y. Patil Biotechnology & Bioinformatics Institute , Pune , India
| | - Pritish Nilendu
- a Cancer and Translational Research Lab , Dr. D.Y. Patil Biotechnology & Bioinformatics Institute , Pune , India
| | - Nilesh Kumar Sharma
- a Cancer and Translational Research Lab , Dr. D.Y. Patil Biotechnology & Bioinformatics Institute , Pune , India
| |
Collapse
|
28
|
Targeting the CXCR4/CXCL12 axis with the peptide antagonist E5 to inhibit breast tumor progression. Signal Transduct Target Ther 2017; 2:17033. [PMID: 29263923 PMCID: PMC5661635 DOI: 10.1038/sigtrans.2017.33] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/14/2017] [Accepted: 04/25/2017] [Indexed: 12/30/2022] Open
Abstract
Emerging evidence has demonstrated that stromal cell-derived factor 1 (SDF-1) and its cognate receptor CXCR4 have critical roles in tumorigenesis, angiogenesis and metastasis. In this study, we demonstrated the significant inhibitory effects of a novel chemically synthetic peptide (E5) on the CXCR4/CXCL12 axis in breast cancer both in vitro and in vivo. E5 was capable of specifically binding to the murine breast cancer cell line 4T1, remarkably inhibiting CXCL12- or stromal cell (MS-5)-induced migration, and adhesion and sensitizing 4T1 cells to multiple chemotherapeutic drugs. Furthermore, E5 combined with either paclitaxel or cyclophosphamide significantly inhibited tumor growth in a breast cancer model. Mechanistic studies implied that E5 can inhibit the expression of CXCR4 to block the CXCL12-mediated recruitment of endothelial progenitor cells and repress CXCR4 downstream of the Akt and Erk signaling pathway, which are involved in tumor angiogenesis and progression. Further pharmacokinetic evaluation suggested that E5 has an acceptable stability, with a half-life of 10 h in healthy mice. In conclusion, E5 demonstrates a promising anti-tumor effect and could be a potential chemotherapeutic sensitizer to improve current clinical breast cancer therapies.
Collapse
|
29
|
Sleightholm RL, Neilsen BK, Li J, Steele MM, Singh RK, Hollingsworth MA, Oupicky D. Emerging roles of the CXCL12/CXCR4 axis in pancreatic cancer progression and therapy. Pharmacol Ther 2017; 179:158-170. [PMID: 28549596 DOI: 10.1016/j.pharmthera.2017.05.012] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chemokine networks regulate a variety of cellular, physiological, and immune processes. These normal functions can become appropriated by cancer cells to facilitate a more hospitable niche for aberrant cells by enhancing growth, proliferation, and metastasis. This is especially true in pancreatic cancer, where chemokine signaling is a vital component in the development of the supportive tumor microenvironment and the signaling between the cancer cells and surrounding stromal cells. Although expression patterns vary among cancer types, the chemokine receptor CXCR4 has been implicated in nearly every major malignancy and plays a prominent role in pancreatic cancer development and progression. This receptor, in conjunction with its primary chemokine ligand CXCL12, promotes pancreatic cancer development, invasion, and metastasis through the management of the tumor microenvironment via complex crosstalk with other pathways. Thus, CXCR4 likely contributes to the poor prognoses observed in patients afflicted with this malignancy. Recent exploration of combination therapies with CXCR4 antagonists have demonstrated improved outcomes, and abolishing the contribution of this pathway may prove crucial to effectively treat pancreatic cancer at both the primary tumor and metastases.
Collapse
Affiliation(s)
- Richard L Sleightholm
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, USA
| | - Beth K Neilsen
- Eppley Institute, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, NE, USA
| | - Jing Li
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, USA
| | - Maria M Steele
- Eppley Institute, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, NE, USA
| | - Rakesh K Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, USA
| | - Michael A Hollingsworth
- Eppley Institute, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, NE, USA; Department of Pathology and Microbiology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, USA
| | - David Oupicky
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
30
|
Tassi RA, Todeschini P, Siegel ER, Calza S, Cappella P, Ardighieri L, Cadei M, Bugatti M, Romani C, Bandiera E, Zanotti L, Tassone L, Guarino D, Santonocito C, Capoluongo ED, Beltrame L, Erba E, Marchini S, D'Incalci M, Donzelli C, Santin AD, Pecorelli S, Sartori E, Bignotti E, Odicino F, Ravaggi A. FOXM1 expression is significantly associated with chemotherapy resistance and adverse prognosis in non-serous epithelial ovarian cancer patients. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:63. [PMID: 28482906 PMCID: PMC5422964 DOI: 10.1186/s13046-017-0536-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/19/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is a spectrum of different diseases, which makes their treatment a challenge. Forkhead box M1 (FOXM1) is an oncogene aberrantly expressed in many solid cancers including serous EOC, but its role in non-serous EOCs remains undefined. We examined FOXM1 expression and its correlation to prognosis across the three major EOC subtypes, and its role in tumorigenesis and chemo-resistance in vitro. METHODS Gene signatures were generated by microarray for 14 clear-cell and 26 endometrioid EOCs, and 15 normal endometrium snap-frozen biopsies. Validation of FOXM1 expression was performed by RT-qPCR and immunohistochemistry in the same samples and additionally in 50 high-grade serous EOCs and in their most adequate normal controls (10 luminal fallopian tube and 20 ovarian surface epithelial brushings). Correlations of FOXM1 expression to clinic-pathological parameters and patients' prognosis were evaluated by Kaplan-Meier and Cox proportional-hazards analyses. OVCAR-3 and two novel deeply characterized EOC cell lines (EOC-CC1 and OSPC2, with clear-cell and serous subtype, respectively) were employed for in vitro studies. Effects of FOXM1 inhibition by transient siRNA transfection were evaluated on cell-proliferation, cell-cycle, colony formation, invasion, and response to conventional first- and second-line anticancer agents, and to the PARP-inhibitor olaparib. Gene signatures of FOXM1-silenced cell lines were generated by microarray and confirmed by RT-qPCR. RESULTS A significant FOXM1 mRNA up-regulation was found in EOCs compared to normal controls. FOXM1 protein overexpression significantly correlated to serous histology (p = 0.001) and advanced FIGO stage (p = 0.004). Multivariate analyses confirmed FOXM1 protein overexpression as an independent indicator of worse disease specific survival in non-serous EOCs, and of shorter time to progression in platinum-resistant cases. FOXM1 downregulation in EOC cell lines inhibited cell growth and clonogenicity, and promoted the cytotoxic effects of platinum compounds, doxorubicin hydrochloride and olaparib. Upon FOXM1 knock-down in EOC-CC1 and OSPC2 cells, microarray and RT-qPCR analyses revealed the deregulation of several common and other unique subtype-specific FOXM1 putative targets involved in cell cycle, metastasis, DNA repair and drug response. CONCLUSIONS FOXM1 is up-regulated in all three major EOCs subtypes, and is a prognostic biomarker and a potential combinatorial therapeutic target in platinum resistant disease, irrespective of tumor histology.
Collapse
Affiliation(s)
- Renata A Tassi
- Department of Obstetrics and Gynecology, "Angelo Nocivelli" Institute of Molecular Medicine, University of Brescia, Brescia, Italy.
| | - Paola Todeschini
- Department of Obstetrics and Gynecology, "Angelo Nocivelli" Institute of Molecular Medicine, University of Brescia, Brescia, Italy
| | - Eric R Siegel
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Stefano Calza
- Department of Molecular and Translational Medicine, Unit of Biostatistics, University of Brescia, Brescia, Italy
| | | | - Laura Ardighieri
- Department of Molecular and Translational Medicine, Section of Pathology, University-ASST Spedali Civili of Brescia, Brescia, Italy
| | - Moris Cadei
- Department of Molecular and Translational Medicine, Section of Pathology, University-ASST Spedali Civili of Brescia, Brescia, Italy
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, Section of Pathology, University-ASST Spedali Civili of Brescia, Brescia, Italy
| | - Chiara Romani
- Department of Obstetrics and Gynecology, "Angelo Nocivelli" Institute of Molecular Medicine, University of Brescia, Brescia, Italy
| | - Elisabetta Bandiera
- Department of Obstetrics and Gynecology, "Angelo Nocivelli" Institute of Molecular Medicine, University of Brescia, Brescia, Italy
| | - Laura Zanotti
- Department of Obstetrics and Gynecology, "Angelo Nocivelli" Institute of Molecular Medicine, University of Brescia, Brescia, Italy
| | - Laura Tassone
- Department of Obstetrics and Gynecology, "Angelo Nocivelli" Institute of Molecular Medicine, University of Brescia, Brescia, Italy
| | - Donatella Guarino
- Laboratory of Clinical Molecular and Personalized Diagnostics, Institute of Biochemistry and Clinical Biochemistry, Catholic University and Foundation Gemelli Hospital, Rome, Italy
| | - Concetta Santonocito
- Laboratory of Clinical Molecular and Personalized Diagnostics, Institute of Biochemistry and Clinical Biochemistry, Catholic University and Foundation Gemelli Hospital, Rome, Italy
| | - Ettore D Capoluongo
- Laboratory of Clinical Molecular and Personalized Diagnostics, Institute of Biochemistry and Clinical Biochemistry, Catholic University and Foundation Gemelli Hospital, Rome, Italy
| | - Luca Beltrame
- Department of Oncology, IRCCS - "Mario Negri" Institute for Pharmacological Research, Milan, Italy
| | - Eugenio Erba
- Department of Oncology, IRCCS - "Mario Negri" Institute for Pharmacological Research, Milan, Italy
| | - Sergio Marchini
- Department of Oncology, IRCCS - "Mario Negri" Institute for Pharmacological Research, Milan, Italy
| | - Maurizio D'Incalci
- Department of Oncology, IRCCS - "Mario Negri" Institute for Pharmacological Research, Milan, Italy
| | - Carla Donzelli
- Department of Molecular and Translational Medicine, Section of Pathology, University-ASST Spedali Civili of Brescia, Brescia, Italy
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Sergio Pecorelli
- Department of Obstetrics and Gynecology, "Angelo Nocivelli" Institute of Molecular Medicine, University of Brescia, Brescia, Italy
| | - Enrico Sartori
- Department of Obstetrics and Gynecology, University of Brescia, Brescia, Italy
| | - Eliana Bignotti
- Division of Obstetrics and Gynecology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Franco Odicino
- Department of Obstetrics and Gynecology, University of Brescia, Brescia, Italy
| | - Antonella Ravaggi
- Department of Obstetrics and Gynecology, "Angelo Nocivelli" Institute of Molecular Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
31
|
Li L, Cai S, Liu S, Feng H, Zhang J. Bioinformatics analysis to screen the key prognostic genes in ovarian cancer. J Ovarian Res 2017; 10:27. [PMID: 28407786 PMCID: PMC5390356 DOI: 10.1186/s13048-017-0323-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/03/2017] [Indexed: 01/21/2023] Open
Abstract
Background Ovarian cancer (OC) is a gynecological oncology that has a poor prognosis and high mortality. This study is conducted to identify the key genes implicated in the prognosis of OC by bioinformatic analysis. Methods Gene expression data (including 568 primary OC tissues, 17 recurrent OC tissues, and 8 adjacent normal tissues) and the relevant clinical information of OC patients were downloaded from The Cancer Genome Atlas database. After data preprocessing, cluster analysis was conducted using the ConsensusClusterPlus package in R. Using the limma package in R, differential analysis was performed to identify feature genes. Based on Kaplan-Meier (KM) survival analysis, prognostic seed genes were selected from the feature genes. After key prognostic genes were further screened by cluster analysis and KM survival analysis, they were performed functional enrichment analysis and multivariate survival analysis. Using the survival package in R, cox regression analysis was conducted for the microarray data of GSE17260 to validate the key prognostic genes. Results A total of 3668 feature genes were obtained, among which 75 genes were identified as prognostic seed genes. Then, 25 key prognostic genes were screened, including AXL, FOS, KLF6, WDR77, DUSP1, GADD45B, and SLIT3. Especially, AXL and SLIT3 were enriched in ovulation cycle. Multivariate survival analysis showed that the key prognostic genes could effectively differentiate the samples and were significantly associated with prognosis. Additionally, GSE17260 confirmed that the key prognostic genes were associated with the prognosis of OC. Conclusion AXL, FOS, KLF6, WDR77, DUSP1, GADD45B, and SLIT3 might affect the prognosis of OC.
Collapse
Affiliation(s)
- Li Li
- The Department of Obstetrics and Gynecology, First Affiliated Hospital, Second Military Medical University, Changhai Road No 168, Shanghai, 200433, People's Republic of China
| | - Shengyun Cai
- The Department of Obstetrics and Gynecology, First Affiliated Hospital, Second Military Medical University, Changhai Road No 168, Shanghai, 200433, People's Republic of China
| | - Shengnan Liu
- The Department of Obstetrics and Gynecology, First Affiliated Hospital, Second Military Medical University, Changhai Road No 168, Shanghai, 200433, People's Republic of China
| | - Hao Feng
- The Department of Obstetrics and Gynecology, First Affiliated Hospital, Second Military Medical University, Changhai Road No 168, Shanghai, 200433, People's Republic of China
| | - Junjie Zhang
- The Department of Obstetrics and Gynecology, First Affiliated Hospital, Second Military Medical University, Changhai Road No 168, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
32
|
Chemotherapy induces adaptive drug resistance and metastatic potentials via phenotypic CXCR4-expressing cell state transition in ovarian cancer. PLoS One 2017; 12:e0171044. [PMID: 28196146 PMCID: PMC5308810 DOI: 10.1371/journal.pone.0171044] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 01/13/2017] [Indexed: 12/22/2022] Open
Abstract
Ovarian cancer (OVC) patients who receive chemotherapy often acquire drug resistance within one year. This can lead to tumor reoccurrence and metastasis, the major causes of mortality. We report a transient increase of a small distinctive CXCR4High/CD24Low cancer stem cell population (CXCR4High) in A2780 and SKOV-3 OVC cell lines in response to cisplatin, doxorubicin, and paclitaxel, treatments. The withdrawal of the drug challenges reversed this cell-state transition. CXCR4High exhibits dormancy in drug resistance and mesenchymal-like invasion, migration, colonization, and tumor formation properties. The removal of this cell population from a doxorubicin-resistant A2780 lineage (A2780/ADR) recovered the sensitivity to drug treatments. A cytotoxic peptide (CXCR4-KLA) that can selectively target cell-surface CXCR4 receptor was further synthesized to investigate the therapeutic merits of targeting CXCR4High. This peptide was more potent than the conventional CXCR4 antagonists (AMD3100 and CTCE-9908) in eradicating the cancer stem cells. When used together with cytotoxic agents such as doxorubicin and cisplatin, the combined drug-peptide regimens exhibited a synergistic cell-killing effect on A2780, A2780/ADR, and SKOV-3. Our data suggested that chemotherapy could establish drug-resistant and tumor-initiating properties of OVC via reversible CXCR4 cell state transition. Therapeutic strategies designed to eradicate rather than antagonize CXCR4High might offer a far-reaching potential as supportive chemotherapy.
Collapse
|
33
|
Xie S, Tu Z, Xiong J, Kang G, Zhao L, Hu W, Tan H, Tembo KM, Ding Q, Deng X, Huang J, Zhang Q. CXCR4 promotes cisplatin-resistance of non-small cell lung cancer in a CYP1B1-dependent manner. Oncol Rep 2016; 37:921-928. [DOI: 10.3892/or.2016.5289] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 10/31/2016] [Indexed: 11/06/2022] Open
|
34
|
Yamabe N, Lee D, Lee H, Shin MS, Hwang GS, Kang KS, Lee JW. Synthesis of Renoprotective Chalcone Analogues That Protect Against Cisplatin-induced Cytotoxicity in LLC-PK1 Cells. B KOREAN CHEM SOC 2016. [DOI: 10.1002/bkcs.10984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Noriko Yamabe
- College of Korean Medicine; Gachon University; Seongnam 461-701 South Korea
| | - Dahae Lee
- College of Korean Medicine; Gachon University; Seongnam 461-701 South Korea
| | - Heesu Lee
- Department of Oral Anatomy, College of Dentistry; Gangneung Wonju National University; Gangneung 210-340 South Korea
| | - Myung Sook Shin
- Natural Constituent Research Center; Korea Institute of Science and Technology; Gangnung 210-340 South Korea
| | - Gwi Seo Hwang
- College of Korean Medicine; Gachon University; Seongnam 461-701 South Korea
| | - Ki Sung Kang
- College of Korean Medicine; Gachon University; Seongnam 461-701 South Korea
| | - Jae Wook Lee
- Natural Constituent Research Center; Korea Institute of Science and Technology; Gangnung 210-340 South Korea
- Convergence Research Center for Dementia; Korea Institute of Science and Technology; Seoul South Korea
- Department of Biological Chemistry; Korea University of Science and Technology (UST); Daejun 305-333 South Korea
| |
Collapse
|
35
|
Kim GY, Park SY, Jo A, Kim M, Leem SH, Jun WJ, Shim SI, Lee SC, Chung JW. Gecko proteins induce the apoptosis of bladder cancer 5637 cells by inhibiting Akt and activating the intrinsic caspase cascade. BMB Rep 2016; 48:531-6. [PMID: 26246284 PMCID: PMC4641238 DOI: 10.5483/bmbrep.2015.48.9.117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Indexed: 01/21/2023] Open
Abstract
Gecko proteins have long been used as anti-tumor agents in oriental medicine, without any scientific background. Although anti-tumor effects of Gecko proteins on several cancers were recently reported, their effect on bladder cancer has not been investigated. Thus, we explored the anti-tumor effect of Gecko proteins and its cellular mechanisms in human bladder cancer 5637 cells. Gecko proteins significantly reduced the viability of 5637 cells without any cytotoxic effect on normal cells. These proteins increased the Annexin-V staining and the amount of condensed chromatin, demonstrating that the Gecko proteinsinduced cell death was caused by apoptosis. Gecko proteins suppressed Akt activation, and the overexpression of constitutively active form of myristoylated Akt prevented Gecko proteins-induced death of 5637 cells. Furthermore, Gecko proteins activated caspase 9 and caspase 3/7. Taken together, our data demonstrated that Gecko proteins suppressed the Akt pathway and activated the intrinsic caspase pathway, leading to the apoptosis of bladder cancer cells. [BMB Reports 2015; 48(9): 531-536].
Collapse
Affiliation(s)
- Geun-Young Kim
- Division of Cardiovascular and Rare Disease, Center for Biomedical Sciences, Korea National Institute of Health, Cheongju 28165, Korea
| | - Soon Yong Park
- Department of Biological Science, Dong-A University, Busan 47315, Korea
| | - Ara Jo
- Department of Biological Science, Dong-A University, Busan 47315, Korea
| | - Mira Kim
- Department of Biological Science, Dong-A University, Busan 47315, Korea
| | - Sun-Hee Leem
- Department of Biological Science, Dong-A University, Busan 47315, Korea
| | - Woo-Jin Jun
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea
| | - Sang In Shim
- Department of Agronomy, Gyeongsang National University, Jinju 52828, Korea
| | - Sang Chul Lee
- Research Center for Integrative Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Jin Woong Chung
- Department of Biological Science, Dong-A University, Busan 47315, Korea
| |
Collapse
|
36
|
Hwang HS, Park IY, Kim DW, Choi SY, Jung YO, Kim HA. PEP-1-FK506BP12 inhibits matrix metalloproteinase expression in human articular chondrocytes and in a mouse carrageenan-induced arthritis model. BMB Rep 2016; 48:407-12. [PMID: 25887750 PMCID: PMC4577291 DOI: 10.5483/bmbrep.2015.48.7.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Indexed: 11/28/2022] Open
Abstract
The 12 kDa FK506-binding protein (FK506BP12), an immunosuppressor, modulates T cell activation via calcineurin inhibition. In this study, we investigated the ability of PEP-1-FK506BP12, consisting of FK506BP12 fused to the protein transduction domain PEP-1 peptide, to suppress catabolic responses in primary human chondrocytes and in a mouse carrageenan-induced paw arthritis model. Western blotting and immunofluorescence analysis showed that PEP-1-FK506BP12 efficiently penetrated chondrocytes and cartilage explants. In interleukin-1β (IL-1β)-treated chondrocytes, PEP-1-FK506BP12 significantly suppressed the expression of catabolic enzymes, including matrix metalloproteinases (MMPs)-1, -3, and -13 in addition to cyclooxygenase-2, at both the mRNA and protein levels, whereas FK506BP12 alone did not. In addition, PEP-1-FK506BP12 decreased IL-1β-induced phosphorylation of the mitogen-activated protein kinase (MAPK) complex (p38, JNK, and ERK) and the inhibitor kappa B alpha. In the mouse model of carrageenan-induced paw arthritis, PEP-1-FK506BP12 suppressed both carrageenan-induced MMP-13 production and paw inflammation. PEP-1-FK506BP12 may have therapeutic potential in the alleviation of OA progression. [BMB Reports 2015; 48(7): 407-412]
Collapse
Affiliation(s)
- Hyun Sook Hwang
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang 431-060; Institute for Skeletal Aging, Hallym University, Chuncheon 200-702, Korea
| | - In Young Park
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang 431-060; Institute for Skeletal Aging, Hallym University, Chuncheon 200-702, Korea
| | - Dae Won Kim
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| | - Young Ok Jung
- Department of Internal Medicine, Kangnam Sacred Heart Hospital, Seoul 150-950, Korea
| | - Hyun Ah Kim
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang 431-060; Institute for Skeletal Aging, Hallym University, Chuncheon 200-702, Korea
| |
Collapse
|
37
|
Choi WT, Yang Y, Xu Y, An J. Targeting chemokine receptor CXCR4 for treatment of HIV-1 infection, tumor progression, and metastasis. Curr Top Med Chem 2016; 14:1574-89. [PMID: 25159167 DOI: 10.2174/1568026614666140827143541] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/30/2014] [Accepted: 06/06/2014] [Indexed: 12/17/2022]
Abstract
The chemokine receptor CXCR4 is required for the entry of human immunodeficiency virus type 1 (HIV-1) into target cells and for the development and dissemination of various types of cancers, including gastrointestinal, cutaneous, head and neck, pulmonary, gynecological, genitourinary, neurological, and hematological malignancies. The T-cell (T)-tropic HIV-1 strains use CXCR4 as the entry coreceptor; consequently, multiple CXCR4 antagonistic inhibitors have been developed for the treatment of acquired immune deficiency syndrome (AIDS). However, other potential applications of CXCR4 antagonists have become apparent since its discovery in 1996. In fact, increasing evidence demonstrates that epithelial and hematopoietic tumor cells exploit the interaction between CXCR4 and its natural ligand, stromal cellderived factor (SDF)-1α, which normally regulates leukocyte migration. The CXCR4 and/or SDF-1α expression patterns in tumor cells also determine the sites of metastatic spread. In addition, the activation of CXCR4 by SDF-1α promotes invasion and proliferation of tumor cells, enhances tumor-associated neoangiogenesis, and assists in the degradation of the extracellular matrix and basement membrane. As such, the evaluation of CXCR4 and/or SDF-1α expression levels has a significant prognostic value in various types of malignancies. Several therapeutic challenges remain to be overcome before the use of CXCR4 inhibitors can be translated into clinical practice, but promising preclinical data demonstrate that CXCR4 antagonists can mobilize tumor cells from their protective microenvironments, interfere with their metastatic and tumorigenic potentials, and/or make tumor cells more susceptible to chemotherapy.
Collapse
Affiliation(s)
| | | | | | - Jing An
- Department of Pharmacology, State University of New York, Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA.
| |
Collapse
|
38
|
Effect of Amino Acids on the Generation of Ginsenoside Rg3 Epimers by Heat Processing and the Anticancer Activities of Epimers in A2780 Human Ovarian Cancer Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:3146402. [PMID: 27051448 PMCID: PMC4804038 DOI: 10.1155/2016/3146402] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/13/2015] [Accepted: 12/03/2015] [Indexed: 12/22/2022]
Abstract
Ginsenosides are the active components of Panax ginseng. Many research studies indicate that these deglycosylated, less-polar ginsenosides have better bioactivity than the major ginsenosides. In the present study, we sought to verify the enhanced anticancer effect of P. ginseng extract after undergoing the Maillard reaction as well as elucidate the underlying mechanism of action. The effects of 9 amino acids were tested; among them, the content of 20(S)-Rg3 in the ginseng extract increased to more than 30, 20, and 20% when processed with valine, arginine, and alanine, respectively, compared with that after normal heat processing. The ginseng extract that was heat-processed with arginine exhibited the most potent inhibitory effect on A2780 ovarian cancer cell proliferation. Therefore, the generation of 20(S)-Rg3 was suggested to be involved in this effect. Moreover, the inhibitory effect of 20(S)-Rg3 on A2780 cell proliferation was significantly stronger than that of 20(R)-Rg3. Protein expression levels of cleaved caspase-3, caspase-8, caspase-9, and PARP in the A2780 ovarian cancer cells markedly increased, whereas the expression of BID decreased after 20(S)-Rg3 treatment. Therefore, we confirmed that the anticancer effects of the products of ginseng that was heat-processed with arginine are mediated mainly via the generation of the less-polar ginsenoside 20(S)-Rg3.
Collapse
|
39
|
Jung K, Lee D, Yu JS, Namgung H, Kang KS, Kim KH. Protective effect and mechanism of action of saponins isolated from the seeds of gac (Momordica cochinchinensis Spreng.) against cisplatin-induced damage in LLC-PK1 kidney cells. Bioorg Med Chem Lett 2016; 26:1466-70. [DOI: 10.1016/j.bmcl.2016.01.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/08/2016] [Accepted: 01/20/2016] [Indexed: 12/14/2022]
|
40
|
CXCR4 over-expression and survival in cancer: a system review and meta-analysis. Oncotarget 2016; 6:5022-40. [PMID: 25669980 PMCID: PMC4467131 DOI: 10.18632/oncotarget.3217] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 12/28/2014] [Indexed: 01/11/2023] Open
Abstract
C-X-C chemokine receptor 4 (CXCR4) is frequently over-expressed in various types of cancer; many agents against CXCR4 are in clinical development currently despite variable data for the prognostic impact of CXCR4 expression. Here eighty-five studies with a total of 11,032 subjects were included to explore the association between CXCR4 and progression-free survival (PFS) or overall survival (OS) in subjects with cancer. Pooled analysis shows that CXCR4 over-expression is significantly associated with poorer PFS (HR 2.04; 95% CI, 1.72-2.42) and OS (HR=1.94; 95% CI, 1.71-2.20) irrespective of cancer types. Subgroup analysis indicates significant association between CXCR4 and shorter PFS in hematological malignancy, breast cancer, colorectal cancer, esophageal cancer, renal cancer, gynecologic cancer, pancreatic cancer and liver cancer; the prognostic effects remained consistent across age, risk of bias, levels of adjustment, median follow-up period, geographical area, detection methods, publication year and size of studies. CXCR4 over-expression predicts unfavorable OS in hematological malignancy, breast cancer, colorectal cancer, esophageal cancer, head and neck cancer, renal cancer, lung cancer, gynecologic cancer, liver cancer, prostate cancer and gallbladder cancer; these effects were independence of age, levels of adjustment, publication year, detection methods and follow-up period. In conclusion, CXCR4 over-expression is associated with poor prognosis in cancer.
Collapse
|
41
|
Kang HR, Lee D, Eom HJ, Lee SR, Lee KR, Kang KS, Kim KH. Identification and mechanism of action of renoprotective constituents from peat moss Sphagnum palustre in cisplatin-induced nephrotoxicity. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.11.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
42
|
Han XG, Du L, Qiao H, Tu B, Wang YG, Qin A, Dai KR, Fan QM, Tang TT. CXCR1 knockdown improves the sensitivity of osteosarcoma to cisplatin. Cancer Lett 2015; 369:405-415. [PMID: 26391645 DOI: 10.1016/j.canlet.2015.09.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/12/2015] [Accepted: 09/08/2015] [Indexed: 12/20/2022]
Abstract
Chemotherapy resistance is a major cause of poor prognoses for osteosarcoma patients. This study aimed to determine whether CXCR1 gene knockdown improves the sensitivity of osteosarcomas to chemotherapy. Both CXCR1 expression and cisplatin sensitivity were investigated and compared in two osteosarcoma cell lines. Sensitivity to the chemotherapy drug cisplatin and apoptosis were investigated with or without stimulation via Interleukin-8 (IL-8), which is a ligand of CXCR1. Furthermore, activation of the Akt signaling pathway was determined. Finally, luciferase-labeled CXCR1-knockdown Saos2-lung cells were injected into the tibiae of nude mice that were treated with cisplatin thereafter. We found that CXCR1 expression and cisplatin sensitivity were negatively correlated in osteosarcoma cell lines. IL-8-induced reduction in sensitivity could be blocked by silencing CXCR1, and CXCR1 knockdown suppressed the Akt signaling pathway. Moreover, CXCR1-knockdown tumors were significantly smaller than control tumors, which was consistent with the luciferase intensity results. The expression levels of IL-8, CXCR1 and p-Akt were suppressed in CXCR1-knockdown cells. Taken together, these data indicate that CXCR1 gene knockdown in osteosarcoma cells improved the sensitivity to chemotherapy and that this process might be regulated in part by the IL-8/CXCR1/Akt signaling pathway.
Collapse
Affiliation(s)
- Xiu-guo Han
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Du
- Department of Orthopedic Surgery, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han Qiao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Tu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-gang Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - An Qin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ke-rong Dai
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi-ming Fan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ting-ting Tang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
43
|
Li XF, Guo XG, Yang YY, Liu AY. Effect of CXCR4 and CD133 co-expression on the prognosis of patients with stage II~III colon cancer. Asian Pac J Cancer Prev 2015; 16:1073-6. [PMID: 25735334 DOI: 10.7314/apjcp.2015.16.3.1073] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To explore the relationship between CXCR4, CD133 co-expression and clinicopathological features as well as prognosis of patients with phase II~III colon cancer. MATERIALS AND METHODS Forty-nine paraffin-embedded samples of tumor tissue and epithelial tissue adjacent to cancer were collected from patients with colon cancer undergoing radical surgery in Baotou Cancer Hospital from January, 2010 to June, 2011. CXCR4 and CD133 expression was detected using immunohistochemistry and its relationship with clinicopathological features and the 3-year survival rate was analyzed. RESULTS In the tumor tissue and colonic epithelial tissue adjacent to cancer, the positive expression rates of CXCR4 were respectively 61.2% (30/49) and 8.16% (4/49), while those of CD133 being 36.7% (18/49) and 6.12% (3/49). CXCR4 and CD133 expression in tumor tissue was not related to patient age, gender, primary focal sites, tumor size, TNM staging, histological type, tumor infiltration depth and presence or absence of lymphatic metastasis, but CXCR4 and CD133 co-expression was associated with TNM staging and lymphatic metastasis. The 3-year survival rate of patients with CXCR4 and CD133 co-expression was 27.3% (3/11), and that of the remainderwas 76.3% (29/38), the difference being significant (χ2=7.0206, p=0.0081). CONCLUSIONS CXCR4 and CD133 co-expression may be a risk factor for poor prognosis of patients with stage II~III colon cancer.
Collapse
Affiliation(s)
- Xiao-Feng Li
- Department of Comprehensive Medical Oncology, Baotou Cancer Hospital, Baotou, China E-mail :
| | | | | | | |
Collapse
|
44
|
Lee S, Jung K, Lee D, Lee SR, Lee KR, Kang KS, Kim KH. Protective effect and mechanism of action of lupane triterpenes from Cornus walteri in cisplatin-induced nephrotoxicity. Bioorg Med Chem Lett 2015; 25:5613-8. [PMID: 26592171 DOI: 10.1016/j.bmcl.2015.10.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/09/2015] [Accepted: 10/13/2015] [Indexed: 01/15/2023]
Abstract
The present study reports a renoprotective effect and the mechanism of action of lupane triterpenes isolated from Cornus walteri in cisplatin-induced renal toxicity. A phytochemical investigation of the MeOH extract of the stems and stem bark of C. walteri resulted in the isolation and identification of twelve lupane triterpenes. Among these, betulinic acid, 29-oxobetulinic acid, betulin 3-acetate, and lupeol ameliorated cisplatin-induced nephrotoxicity to 80% of the control value at 125 μM. Upregulated phosphorylation of JNK, ERK, and p38 following cisplatin treatment were markedly decreased after co-treatment with betulinic acid, 29-oxobetulinic acid, betulin 3-acetate, and lupeol. In addition, the protein expression level of cleaved caspase-3 and the percentage of apoptotic cells were also significantly reduced after co-treatment with betulinic acid, 29-oxobetulinic acid, betulin 3-acetate, and lupeol. These results show that blocking the MAPK signaling cascade plays a critical role in mediating the renoprotective effect of betulinic acid, 29-oxobetulinic acid, betulin 3-acetate, and lupeol isolated from C. walteri extract.
Collapse
Affiliation(s)
- Seulah Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Kiwon Jung
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Seongnam 13488, Republic of Korea
| | - Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Seoung Rak Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Kang Ro Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea.
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea.
| |
Collapse
|
45
|
Protective Effect of Artemisia asiatica Extract and Its Active Compound Eupatilin against Cisplatin-Induced Renal Damage. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:483980. [PMID: 26539226 PMCID: PMC4619882 DOI: 10.1155/2015/483980] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 06/14/2015] [Indexed: 12/20/2022]
Abstract
The present study investigated the renoprotective effect of an Artemisia asiatica extract and eupatilin in kidney epithelial (LLC-PK1) cells. Although cisplatin is effective against several cancers, its use is limited due to severe nephrotoxicity. Eupatilin is a flavonoid compound isolated from the Artemisia plant and possesses antioxidant as well as potent anticancer properties. In the LLC-PK1 cellular model, the decline in cell viability induced by oxidative stress, such as that induced by cisplatin, was significantly and dose-dependently inhibited by the A. asiatica extract and eupatilin. The increased protein expressions of phosphorylated JNK and p38 by cisplatin in cells were markedly reduced after A. asiatica extract or eupatilin cotreatment. The elevated expression of cleaved caspase-3 was significantly reduced by A. asiatica extract and eupatilin, and the elevated percentage of apoptotic cells after cisplatin treatment in LLC-PK1 cells was markedly decreased by cotreatment with A. asiatica extract or eupatilin. Taken together, these results suggest that A. asiatica extract and eupatilin could cure or prevent cisplatin-induced renal toxicity without any adverse effect; thus, it can be used in combination with cisplatin to prevent nephrotoxicity.
Collapse
|
46
|
Yang D, Dai T, Xue L, Liu X, Wu B, Geng J, Mao X, Wang R, Chen L, Chu X. Expression of chemokine receptor CXCR7 in colorectal carcinoma and its prognostic significance. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:13051-13058. [PMID: 26722500 PMCID: PMC4680445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/22/2015] [Indexed: 06/05/2023]
Abstract
Previous studies have shown that chemokine receptor CXCR7 plays critical roles in tumor development. However, the clinicopathological and prognostic significance of CXCR7 in colorectal carcinoma (CRC) has not been fully understood. The aim of our study is to investigate the expression of CXCR7 and its clinical significance in CRC. First, quantitative RT-PCR and Western blot assays were performed to determine the expression of CXCR7 mRNA and protein in 20 paired of CRC tissues and corresponding adjacent non-tumor tissues. Next, immunohistochemistry was performed to detect the expression of CXCR7 protein in another 96 cases of CRC tissues, and analyze its correlation with clinicopathological factors of patients. Finally, the correlation of CXCR7 with 5-year overall survival (OS) and progression free survival (PFS) was statistically analyzed by the Kaplan-Meier method and Cox proportional hazards model. Results showed that the expression levels of CXCR7 mRNA and protein were significantly higher in CRC tissues than in normal tissues. Positive CXCR7 expression was observed to be significantly correlated with lymph nodal metastasis (P < 0.001), distant metastasis (P = 0.017), and advanced TNM stage (P < 0.001). Patients with positive expression of CXCR7 were demonstrated to be associated with worse OS and PFS (P < 0.001, P < 0.001, respectively). Moreover, multivariate survival analysis revealed that CXCR7 expression level might be an independent predictive factor for OS and PFS of CRC patients. Collectively, positive CXCR7 expression in CRC was correlated with tumor development and poor prognosis of patients.
Collapse
Affiliation(s)
- Dan Yang
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical UniversityNanjing, Jiangsu Province, China
| | - Tingting Dai
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical UniversityNanjing, Jiangsu Province, China
| | - Lijun Xue
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical UniversityNanjing, Jiangsu Province, China
| | - Xiaobei Liu
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical UniversityNanjing, Jiangsu Province, China
| | - Bo Wu
- Department of Pathology, Jinling Hospital, Nanjing Clinical School of Southern Medical UniversityNanjing, Jiangsu Province, China
| | - Jian Geng
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical UniversityNanjing, Jiangsu Province, China
| | - Xiaobei Mao
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical UniversityNanjing, Jiangsu Province, China
| | - Rui Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing UniversityNanjing, Jiangsu Province, China
| | - Longbang Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing UniversityNanjing, Jiangsu Province, China
| | - Xiaoyuan Chu
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical UniversityNanjing, Jiangsu Province, China
| |
Collapse
|
47
|
Lee MH, Cho Y, Jung BC, Kim SH, Kang YW, Pan CH, Rhee KJ, Kim YS. Parkin induces G2/M cell cycle arrest in TNF-α-treated HeLa cells. Biochem Biophys Res Commun 2015; 464:63-9. [DOI: 10.1016/j.bbrc.2015.05.101] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 05/29/2015] [Indexed: 10/23/2022]
|
48
|
Han MS, Han IH, Lee D, An JM, Kim SN, Shin MS, Yamabe N, Hwang GS, Yoo HH, Choi SJ, Kang KS, Jang HJ. Beneficial effects of fermented black ginseng and its ginsenoside 20(S)-Rg3 against cisplatin-induced nephrotoxicity in LLC-PK1 cells. J Ginseng Res 2015; 40:135-40. [PMID: 27158234 PMCID: PMC4845053 DOI: 10.1016/j.jgr.2015.06.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 01/23/2023] Open
Abstract
Background Nephrotoxicity is a common side effect of medications. Panax ginseng is one of the best-known herbal medicines, and its individual constituents enhance renal function. Identification of its efficacy and mechanisms of action against drug-induced nephrotoxicity, as well as the specific constituents mediating this effect, have recently emerged as an interesting research area focusing on the kidney protective efficacy of P. ginseng. Methods The present study investigated the kidney protective effect of fermented black ginseng (FBG) and its active component ginsenoside 20(S)-Rg3 against cisplatin (chemotherapy drug)-induced damage in pig kidney (LLC-PK1) cells. It focused on assessing the role of mitogen-activated protein kinases as important mechanistic elements in kidney protection. Results The reduced cell viability induced by cisplatin was significantly recovered with FBG extract and ginsenoside 20(S)-Rg3 dose-dependently. The cisplatin-induced elevated protein levels of phosphorylated c-Jun N-terminal kinase (JNK), p53, and cleaved caspase-3 were decreased after cotreatment with FBG extract or ginsenoside 20(S)-Rg3. The elevated percentage of apoptotic LLC-PK1 cells induced by cisplatin treatment was significantly abrogated by cotreatment with FBG and the ginsenoside 20(S)-Rg3. Conclusion FBG and its major ginsenoside 20(S)-Rg3, ameliorated cisplatin-induced nephrotoxicity in LLC-PK1 cells by blocking the JNK–p53–caspase-3 signaling cascade.
Collapse
Affiliation(s)
- Myoung-Sik Han
- Department of Surgery, University of Ulsan College of Medicine, Gangneung, Korea
| | - Im-Ho Han
- Department of Chemistry, Gangneung-Wonju National University, Gangneung, Korea; Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung, Korea
| | - Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam, Korea
| | - Jun Min An
- GINSENG BY PHARM Co., Ltd., Wonju, Korea
| | - Su-Nam Kim
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung, Korea
| | - Myoung-Sook Shin
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung, Korea
| | - Noriko Yamabe
- College of Korean Medicine, Gachon University, Seongnam, Korea
| | - Gwi Seo Hwang
- College of Korean Medicine, Gachon University, Seongnam, Korea
| | - Hye Hyun Yoo
- College of Pharmacy, Hanyang University, Ansan, Korea
| | - Suk-Jung Choi
- Department of Chemistry, Gangneung-Wonju National University, Gangneung, Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam, Korea
| | - Hyuk-Jai Jang
- Department of Surgery, University of Ulsan College of Medicine, Gangneung, Korea
| |
Collapse
|
49
|
Park JY, Choi P, Kim T, Ko H, Kim HK, Kang KS, Ham J. Protective Effects of Processed Ginseng and Its Active Ginsenosides on Cisplatin-Induced Nephrotoxicity: In Vitro and in Vivo Studies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:5964-5969. [PMID: 26050847 DOI: 10.1021/acs.jafc.5b00782] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Although cisplatin can dramatically improve the survival rate in cancer patients, its use is limited by its nephrotoxicity. Previous investigations showed that Panax ginseng contains components that exhibit protective activity against cisplatin-induced nephropathy. The aim of the present study is to investigate the effect of microwave-assisted processing on the protective effect of ginseng and identify ginsenosides that are active against cisplatin-induced kidney damage to evaluate the potential of using ginseng in the management of nephrotoxicity. The LLC-PK1 cell damage by cisplatin was significantly decreased by treatment with microwave-processed ginseng (MG) and ginsenosides Rg3, Rg5, and Rk1. Reduced expression of p53 and c-Jun N-terminal kinase proteins by cisplatin in LLC-PK1 cells was markedly ameliorated after Rg3 and Rg5/Rk1 treatment. Additionally, elevated expression of cleaved caspase-3 was significantly reduced by ginsenosides Rg5, Rk1, and with even greater potency, Rg3. Moreover, MG and its fraction containing active ginsenosides showed protective effects against cisplatin-induced nephropathy in mice. We found that ginsenosides Rg3, Rg5, and Rk1 generated during the heat treatment of ginseng ameliorate renal damage by regulating inflammation and apoptosis. Results of current experiments provide evidence of the renoprotective effects and therapeutic potential of MG and its active ginsenosides, both in vitro and in vivo.
Collapse
Affiliation(s)
- Jun Yeon Park
- ‡College of Korean Medicine, Gachon University, Seongnam 461-701, South Korea
| | - Pilju Choi
- §KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology (KIST), Gangneung 210-340, South Korea
| | - Taejung Kim
- §KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology (KIST), Gangneung 210-340, South Korea
| | - Hyeonseok Ko
- ∥Laboratory of Molecular Oncology, Cheil General Hospital and Women's Healthcare Center, Dankook University College of Medicine, Seoul 100-380, South Korea
| | - Ho-kyong Kim
- ⊥Richwood Pharmaceutical Company, Limited, Seoul 100-704, South Korea
| | - Ki Sung Kang
- ‡College of Korean Medicine, Gachon University, Seongnam 461-701, South Korea
| | - Jungyeob Ham
- §KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology (KIST), Gangneung 210-340, South Korea
| |
Collapse
|
50
|
Liang S, Peng X, Li X, Yang P, Xie L, Li Y, Du C, Zhang G. Silencing of CXCR4 sensitizes triple-negative breast cancer cells to cisplatin. Oncotarget 2015; 6:1020-30. [PMID: 25544759 PMCID: PMC4359214 DOI: 10.18632/oncotarget.2741] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 11/08/2014] [Indexed: 02/05/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer for which there is no effective treatment. Previously, we and others demonstrated that CXCR4 surface expression is an independent prognostic factor for disease relapse and survival in breast cancer. In this study, we investigated the effects of CXCR4 gene silencing on cisplatin chemosensitivity in human triple-negative breast cancer cell lines. We found that CXCR4 silencing significantly inhibited cell growth, decreased colony formation, and enhanced cisplatin sensitivity while overexpression of CXCR4 rendered cells more resistant to cisplatin. Moreover, the percentage of apoptosis and cell cycle arrest at the G2/M phase of cisplatin-treated CXCR4 knockdown cells was significantly higher than control cells. Furthermore, we demonstrated CXCR4 knockdown cells showed lower levels of mutant p53 and Bcl-2 protein than the control group, while also having higher levels of caspase-3 and Bax. However overexpression of CXCR4 had the reverse effect. In vivo experiments confirmed that downregulation of CXCR4 enhanced cisplatin anticancer activity in tumor-bearing mice, and that this enhanced anticancer activity is attributable to tumor cell apoptosis. Thus, this study indicates that CXCR4 can modulate cisplatin sensitivity in TNBC cells and suggests that CXCR4 may be a therapeutic target for TNBC.
Collapse
Affiliation(s)
- Sixian Liang
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Xun Peng
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Xiaoli Li
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Ping Yang
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Linhao Xie
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Yaochen Li
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Caiwen Du
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Guojun Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| |
Collapse
|