1
|
Chen W, Feng H, Mo Y, Pan Z, Ji S, Liang H, Shen XC, Jiang BP. Hyaluronic acid-functionalized ruthenium photothermal nanoenzyme for enhancing osteosarcoma chemotherapy: Cascade targeting and bidirectional modulation of drug resistance. Carbohydr Polym 2025; 349:122945. [PMID: 39643406 DOI: 10.1016/j.carbpol.2024.122945] [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: 08/17/2024] [Revised: 10/24/2024] [Accepted: 11/03/2024] [Indexed: 12/09/2024]
Abstract
Insufficient drug delivery efficiency in vivo and robust drug resistance are two major factors to induce suboptimal efficacy in chemotherapy of osteosarcoma (OS). To address these challenges, we developed polysaccharide hyaluronic acid (HA)-functionalized ruthenium nanoaggregates (Ru NAs) to enhance the chemotherapy of doxorubicin (DOX) for OS. These NAs, comprising Ru nanoparticles (NPs) and alendronate-modified HA (HA-ALN), effectively load DOX, resulting in DOX@Ru-HA-ALN NAs. The combination of HA and ALN in NAs ensures outstanding cascade targeting towards tumor-invaded bone tissues and CD44-overexpressing tumor cells, maximizing therapeutic efficacy while minimizing off-target effects. Concurrently, the Ru NPs in NAs function as "smart" photoenzymatic agent to not only in situ relieve hypoxia of OS via the catalysis of overexpressed H2O2 to produce O2, but also generate mild photothermal effect under 808-nm laser irradiation. They can bidirectionally overcome drug resistance of DOX via downregulation of resistance-related factors including multi-drug resistant associate protein, P-glycoprotein, heat shock factor 1, etc. The integration of cascade targeting with bidirectional modulation of drug resistance positions Ru-HA-ALN NAs to substantially enhance DOX chemotherapy for OS. Therefore, the present work highlights the potential of polysaccharide-functionalized nanomaterials in advancing tumor chemotherapy by addressing challenges of both delivery efficiency and drug resistance.
Collapse
Affiliation(s)
- Weifeng Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Hao Feng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Yinyin Mo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Zhihui Pan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Shichen Ji
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Xing-Can Shen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China.
| | - Bang-Ping Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China.
| |
Collapse
|
2
|
Geng S, Zhu L, Wang Y, Liu Q, Yu C, Shi S, Yu S. Co-Colorectal cancer stem cells employ the FADS1/DDA axis to evade NK cell-mediated immunosuppression after co-cultured with NK cells under hypoxia. Int Immunopharmacol 2024; 143:113535. [PMID: 39488917 DOI: 10.1016/j.intimp.2024.113535] [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: 07/01/2024] [Revised: 10/19/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
Colorectal cancer (CRC) ranks as China's second most common cancer and fifth top cancer death cause. The study highlights the role of Natural Killer (NK) cells in targeting cancer stem cells (CSCs) that evade immune responses in CRC. Colorectal cancer stem cells (CCSCs) were stem from HT-29 cells and co-cultured with NK cells under normoxic or hypoxic conditions. The impact of this co-culture was evaluated using CCK8 assays for NK cell viability, ELISA for cytokine level changes, and flow cytometry for assessing NK cell apoptosis and activation. Comprehensive metabolomic and transcriptomic analyses were also performed to identify key genes and metabolites involved in the interaction between CCSCs and NK cells Co-culture of CCSCs with NK cells under hypoxia reduced NK cytotoxicity, increased NK apoptosis, and altered cytokine secretion by decreasing IFN-γ and TNF-α levels while increasing IL-6. Transcriptomic and metabolomic analysis identified 4 genes (FADS1, ALDH3A2, GCSH, MTCL1) and 3 metabolites (glyoxylic acid, spermine, DDA) as significant. Interfering with FADS1 counteracted the suppression of IFN-γ and TNF-α induced by CSC cells. Curiously, this inhibition caused by si-FADS1 could be neutralized by the addition of exogenous DDA. Co-culturing with NK cells notably increased spermine levels. Exogenous spermine resulted in a significant reduction in HT-29 cell death rates at 32 µM, 64 µM, and 128 µM, compared to NK cells without spermine. Our research explored CCSCs employed the FADS1/DDA axis to evade NK cell-mediated immunosuppression after co-cultured with NK cells under hypoxia.
Collapse
Affiliation(s)
- Shan Geng
- Central Laboratory of the People's Hospital of Dazu, The Affiliated Dazu Hospital of Chongqing Medical University, 402360 Chongqing, China
| | - Lei Zhu
- Department of General Surgery, The First People's Hospital of Kunming, 650034 Kunming, Yunnan Province, China
| | - Yanping Wang
- Central Laboratory of the People's Hospital of Dazu, The Affiliated Dazu Hospital of Chongqing Medical University, 402360 Chongqing, China
| | - Qiang Liu
- Department of General Surgery, The Affiliated Dazu Hospital of Chongqing Medical University, 402360 Chongqing, China
| | - Caiyu Yu
- Department of Hernia Surgery, Qujing No.1 Hospital, 655099 Qujing, Yunnan Province, China
| | - Shan Shi
- Office of Hospital, The Affiliated Dazu Hospital of Chongqing Medical University, 402360 Chongqing, China.
| | - Shaohong Yu
- Department of General Surgery, The Affiliated Dazu Hospital of Chongqing Medical University, 402360 Chongqing, China.
| |
Collapse
|
3
|
Zhou Y, Na C, Li Z. Novel insights into immune cells modulation of tumor resistance. Crit Rev Oncol Hematol 2024; 202:104457. [PMID: 39038527 DOI: 10.1016/j.critrevonc.2024.104457] [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: 01/19/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024] Open
Abstract
Tumor resistance poses a significant challenge to effective cancer treatment, making it imperative to explore new therapeutic strategies. Recent studies have highlighted the profound involvement of immune cells in the development of tumor resistance. Within the tumor microenvironment, macrophages undergo polarization into the M2 phenotype, thus promoting the emergence of drug-resistant tumors. Neutrophils contribute to tumor resistance by forming extracellular traps. While T cells and natural killer (NK) cells exert their impact through direct cytotoxicity against tumor cells. Additionally, dendritic cells (DCs) have been implicated in preventing tumor drug resistance by stimulating T cell activation. In this review, we provide a comprehensive summary of the current knowledge regarding immune cell-mediated modulation of tumor resistance at the molecular level, with a particular focus on macrophages, neutrophils, DCs, T cells, and NK cells. The targeting of immune cell modulation exhibits considerable potential for addressing drug resistance, and an in-depth understanding of the molecular interactions between immune cells and tumor cells holds promise for the development of innovative therapies. Furthermore, we explore the clinical implications of these immune cells in the treatment of drug-resistant tumors. This review emphasizes the exploration of novel approaches that harness the functional capabilities of immune cells to effectively overcome drug-resistant tumors.
Collapse
Affiliation(s)
- Yi Zhou
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Chuhan Na
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Zhigang Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen 518107, China.
| |
Collapse
|
4
|
Wu D, Li X, Khan FA, Yuan C, Pandupuspitasari NS, Huang C, Sun F, Guan K. tRNA modifications and tRNA-derived small RNAs: new insights of tRNA in human disease. Cell Biol Toxicol 2024; 40:76. [PMID: 39276283 PMCID: PMC11401796 DOI: 10.1007/s10565-024-09919-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/04/2024] [Indexed: 09/16/2024]
Abstract
tRNAs are codon decoders that convert the transcriptome into the proteome. The field of tRNA research is excited by the increasing discovery of specific tRNA modifications that are installed at specific, evolutionarily conserved positions by a set of specialized tRNA-modifying enzymes and the biogenesis of tRNA-derived regulatory fragments (tsRNAs) which exhibit copious activities through multiple mechanisms. Dysregulation of tRNA modification usually has pathological consequences, a phenomenon referred to as "tRNA modopathy". Current evidence suggests that certain tRNA-modifying enzymes and tsRNAs may serve as promising diagnostic biomarkers and therapeutic targets, particularly for chemoresistant cancers. In this review, we discuss the latest discoveries that elucidate the molecular mechanisms underlying the functions of clinically relevant tRNA modifications and tsRNAs, with a focus on malignancies. We also discuss the therapeutic potential of tRNA/tsRNA-based therapies, aiming to provide insights for the development of innovative therapeutic strategies. Further efforts to unravel the complexities inherent in tRNA biology hold the promise of yielding better biomarkers for the diagnosis and prognosis of diseases, thereby advancing the development of precision medicine for health improvement.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Xiuling Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Faheem Ahmed Khan
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | - Chenyang Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | | | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
5
|
Adams KM, Wendt JR, Wood J, Olson S, Moreno R, Jin Z, Gopalan S, Lang JD. Cell-intrinsic platinum response and associated genetic and gene expression signatures in ovarian cancer cell lines and isogenic models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605381. [PMID: 39131380 PMCID: PMC11312449 DOI: 10.1101/2024.07.26.605381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Ovarian cancers are still largely treated with platinum-based chemotherapy as the standard of care, yet few biomarkers of clinical response have had an impact on clinical decision making as of yet. Two particular challenges faced in mechanistically deciphering platinum responsiveness in ovarian cancer have been the suitability of cell line models for ovarian cancer subtypes and the availability of information on comparatively how sensitive ovarian cancer cell lines are to platinum. We performed one of the most comprehensive profiles to date on 36 ovarian cancer cell lines across over seven subtypes and integrated drug response and multiomic data to improve on our understanding of the best cell line models for platinum responsiveness in ovarian cancer. RNA-seq analysis of the 36 cell lines in a single batch experiment largely conforms with the currently accepted subtyping of ovarian cancers, further supporting other studies that have reclassified cell lines and demonstrate that commonly used cell lines are poor models of high-grade serous ovarian carcinoma. We performed drug dose response assays in the 32 of these cell lines for cisplatin and carboplatin, providing a quantitative database of IC50s for these drugs. Our results demonstrate that cell lines largely fall either well above or below the equivalent dose of the clinical maximally achievable dose (Cmax) of each compound, allowing designation of cell lines as sensitive or resistant. We performed differential expression analysis for high-grade serous ovarian carcinoma cell lines to identify gene expression correlating with platinum-response. Further, we generated two platinum-resistant derivatives each for OVCAR3 and OVCAR4, as well as leveraged clinically-resistant PEO1/PEO4/PEO6 and PEA1/PEA2 isogenic models to perform differential expression analysis for seven total isogenic pairs of platinum resistant cell lines. While gene expression changes overall were heterogeneous and vast, common themes were innate immunity/STAT activation, epithelial to mesenchymal transition and stemness, and platinum influx/efflux regulators. In addition to gene expression analyses, we performed copy number signature analysis and orthogonal measures of homologous recombination deficiency (HRD) scar scores and copy number burden, which is the first report to our knowledge applying field-standard copy number signatures to ovarian cancer cell lines. We also examined markers and functional readouts of stemness that revealed that cell lines are poor models for examination of stemness contributions to platinum resistance, likely pointing to the fact that this is a transient state. Overall this study serves as a resource to determine the best cell lines to utilize for ovarian cancer research on certain subtypes and platinum response studies, as well as sparks new hypotheses for future study in ovarian cancer.
Collapse
Affiliation(s)
- Kristin M. Adams
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Jae-Rim Wendt
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Josie Wood
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Sydney Olson
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Moreno
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Department of Computer Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhongmou Jin
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Srihari Gopalan
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Jessica D. Lang
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
6
|
Kaur K, Sanghu J, Memarzadeh S, Jewett A. Exploring the Potential of Natural Killer Cell-Based Immunotherapy in Targeting High-Grade Serous Ovarian Carcinomas. Vaccines (Basel) 2024; 12:677. [PMID: 38932405 PMCID: PMC11209217 DOI: 10.3390/vaccines12060677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/05/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
High-grade serous ovarian cancers (HGSOCs) likely consist of poorly differentiated stem-like cells (PDSLCs) and differentiated tumor cells. Conventional therapeutics are incapable of completely eradicating PDSLCs, contributing to disease progression and tumor relapse. Primary NK cells are known to effectively lyse PDSLCs, but they exhibit low or minimal cytotoxic potential against well-differentiated tumors. We have introduced and discussed the characteristics of super-charged NK (sNK) cells in this review. sNK cells, in comparison to primary NK cells, exhibit a significantly higher capability for the direct killing of both PDSLCs and well-differentiated tumors. In addition, sNK cells secrete significantly higher levels of cytokines, especially those known to induce the differentiation of tumors. In addition, we propose that a combination of sNK and chemotherapy could be one of the most effective strategies to eliminate the heterogeneous population of ovarian tumors; sNK cells can lyse both PDSLCs and well-differentiated tumors, induce the differentiation of PDSLCs, and could be used in combination with chemotherapy to target both well-differentiated and NK-induced differentiated tumors.
Collapse
Affiliation(s)
- Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA 90095, USA;
| | - Jashan Sanghu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; (J.S.); (S.M.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sanaz Memarzadeh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; (J.S.); (S.M.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- The Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
- The VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA 90095, USA;
- The Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
7
|
Geethadevi A, Ku Z, Tsaih SW, Parashar D, Kadamberi IP, Xiong W, Deng H, George J, Kumar S, Mittal S, Zhang N, Pradeep S, An Z, Chaluvally-Raghavan P. Blocking Oncostatin M receptor abrogates STAT3 mediated integrin signaling and overcomes chemoresistance in ovarian cancer. NPJ Precis Oncol 2024; 8:127. [PMID: 38839865 PMCID: PMC11153533 DOI: 10.1038/s41698-024-00593-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Chemotherapy such as cisplatin is widely used to treat ovarian cancer either before or after surgical debulking. However, cancer relapse due to chemotherapy resistance is a major challenge in the treatment of ovarian cancer. The underlying mechanisms related to chemotherapy resistance remain largely unclear. Therefore, identification of effective therapeutic strategies is urgently needed to overcome therapy resistance. Transcriptome-based analysis, in vitro studies and functional assays identified that cisplatin-resistant ovarian cancer cells express high levels of OSMR compared to cisplatin sensitive cells. Furthermore, OSMR expression associated with a module of integrin family genes and predominantly linked with integrin αV (ITGAV) and integrin β3 (ITGB3) for cisplatin resistance. Using ectopic expression and knockdown approaches, we proved that OSMR directly regulates ITGAV and ITGB3 gene expression through STAT3 activation. Notably, targeting OSMR using anti-OSMR human antibody inhibited the growth and metastasis of ovarian cancer cells and sensitized cisplatin treatment. Taken together, our results underscore the pivotal role of OSMR as a requirement for cisplatin resistance in ovarian cancer. Notably, OSMR fostered the expression of a distinct set of integrin genes, which in turn resulted into a crosstalk between OSMR and integrins for signaling activation that is critical for cisplatin resistance. Therefore, targeting OSMR emerges as a promising and viable strategy to reverse cisplatin-resistance in ovarian cancer.
Collapse
Affiliation(s)
- Anjali Geethadevi
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Shirng-Wern Tsaih
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Medicine, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ishaque P Kadamberi
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Hui Deng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Jasmine George
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sudhir Kumar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sonam Mittal
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Sunila Pradeep
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA.
| | - Pradeep Chaluvally-Raghavan
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA.
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
8
|
Carmi YK, Agbarya A, Khamaisi H, Farah R, Shechtman Y, Korobochka R, Gopas J, Mahajna J. Ovarian cancer ascites confers platinum chemoresistance to ovarian cancer cells. Transl Oncol 2024; 44:101939. [PMID: 38489872 PMCID: PMC10955424 DOI: 10.1016/j.tranon.2024.101939] [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: 09/21/2023] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 03/17/2024] Open
Abstract
Ovarian cancer (OC), the second most common form of gynecologic malignancy, has a poor prognosis and is often discovered in the late stages. Platinum-based chemotherapy is the first line of therapy. Nevertheless, treatment OC has proven challenging due to toxicity and the development of acquired resistance to therapy. Tumor microenvironment (TME) has been associated with platinum chemoresistance. Malignant ascites has been used as OC tumor microenvironment and its ability to induce platinum chemoresistance has been investigated. Our results suggest that exposure to OC ascites induces platinum chemoresistance in 11 of 13 cases (85 %) on OC cells. In contrast, 75 % of cirrhotic ascites (3 of 4) failed to confer platinum chemoresistance to OC cells. Cytokine array analysis revealed that IL -6 and to a lesser extent HGF were enriched in OC ascites, whereas IL -22 was enriched in cirrhotic ascites. Pharmaceutical inhibitors targeting the IL -6/ JAK pathway were mildly effective in overcoming platinum chemoresistance induced by malignant ascites. In contrast, crizotinib, an HGF/c- MET inhibitor, and 2-hydroxyestradiol (2HE2) were effective in restoring platinum chemosensitivity to OC. Our results demonstrate the importance of OC ascites in supporting platinum chemoresistance and the potential of combination therapy to restore chemosensitivity of OC cells.
Collapse
Affiliation(s)
- Yifat Koren Carmi
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel; Shraga Segal Department of Microbiology, Immunology and Genetics, and Department of Oncology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Abed Agbarya
- Oncology Department, Bnai Zion MC, Haifa, Israel
| | - Hazem Khamaisi
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel
| | - Raymond Farah
- Department of Internal Medicine, Ziv Medical Center, Safed, Israel
| | | | | | - Jacob Gopas
- Shraga Segal Department of Microbiology, Immunology and Genetics, and Department of Oncology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Jamal Mahajna
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel; Department of Biotechnology, Tel-Hai College, Kiryat Shmona, Israel.
| |
Collapse
|
9
|
Schaff DL, Fasse AJ, White PE, Vander Velde RJ, Shaffer SM. Clonal differences underlie variable responses to sequential and prolonged treatment. Cell Syst 2024; 15:213-226.e9. [PMID: 38401539 PMCID: PMC11003565 DOI: 10.1016/j.cels.2024.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 11/14/2023] [Accepted: 01/29/2024] [Indexed: 02/26/2024]
Abstract
Cancer cells exhibit dramatic differences in gene expression at the single-cell level, which can predict whether they become resistant to treatment. Treatment perpetuates this heterogeneity, resulting in a diversity of cell states among resistant clones. However, it remains unclear whether these differences lead to distinct responses when another treatment is applied or the same treatment is continued. In this study, we combined single-cell RNA sequencing with barcoding to track resistant clones through prolonged and sequential treatments. We found that cells within the same clone have similar gene expression states after multiple rounds of treatment. Moreover, we demonstrated that individual clones have distinct and differing fates, including growth, survival, or death, when subjected to a second treatment or when the first treatment is continued. By identifying gene expression states that predict clone survival, this work provides a foundation for selecting optimal therapies that target the most aggressive resistant clones within a tumor. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Dylan L Schaff
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19146, USA
| | - Aria J Fasse
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19146, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Phoebe E White
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19146, USA
| | - Robert J Vander Velde
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19146, USA; Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19146, USA
| | - Sydney M Shaffer
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19146, USA; Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19146, USA.
| |
Collapse
|
10
|
Jung J, Han H. The diverse influences of relaxin-like peptide family on tumor progression: Potential opportunities and emerging challenges. Heliyon 2024; 10:e24463. [PMID: 38298643 PMCID: PMC10828710 DOI: 10.1016/j.heliyon.2024.e24463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 02/02/2024] Open
Abstract
Relaxin-like peptide family exhibit differential expression patterns in various types of cancers and play a role in cancer development. This family participates in tumorigenic processes encompassing proliferation, migration, invasion, tumor microenvironment, immune microenvironment, and anti-cancer resistance, ultimately influencing patient prognosis. In this review, we explore the mechanisms underlying the interaction between the RLN-like peptide family and tumors and provide an overview of therapeutic approaches utilizing this interaction.
Collapse
Affiliation(s)
| | - Hyunho Han
- Department of Urology, Urological Science Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
11
|
Wilczyński J, Paradowska E, Wilczyńska J, Wilczyński M. Prediction of Chemoresistance-How Preclinical Data Could Help to Modify Therapeutic Strategy in High-Grade Serous Ovarian Cancer. Curr Oncol 2023; 31:229-249. [PMID: 38248100 PMCID: PMC10814576 DOI: 10.3390/curroncol31010015] [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: 11/13/2023] [Revised: 12/12/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is one of the most lethal tumors generally and the most fatal cancer of the female genital tract. The approved standard therapy consists of surgical cytoreduction and platinum/taxane-based chemotherapy, and of targeted therapy in selected patients. The main therapeutic problem is chemoresistance of recurrent and metastatic HGSOC tumors which results in low survival in the group of FIGO III/IV. Therefore, the prediction and monitoring of chemoresistance seems to be of utmost importance for the improvement of HGSOC management. This type of cancer has genetic heterogeneity with several subtypes being characterized by diverse gene signatures and disturbed peculiar epigenetic regulation. HGSOC develops and metastasizes preferentially in the specific intraperitoneal environment composed mainly of fibroblasts, adipocytes, and immune cells. Different HGSOC subtypes could be sensitive to distinct sets of drugs. Moreover, primary, metastatic, and recurrent tumors are characterized by an individual biology, and thus diverse drug responsibility. Without a precise identification of the tumor and its microenvironment, effective treatment seems to be elusive. This paper reviews tumor-derived genomic, mutational, cellular, and epigenetic biomarkers of HGSOC drug resistance, as well as tumor microenvironment-derived biomarkers of chemoresistance, and discusses their possible use in the novel complex approach to ovarian cancer therapy and monitoring.
Collapse
Affiliation(s)
- Jacek Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland;
| | - Justyna Wilczyńska
- Department of Tele-Radiotherapy, Mikolaj Kopernik Provincial Multi-Specialized Oncology and Traumatology Center, 62 Pabianicka Str., 93-513 Lodz, Poland;
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother’s Health Center—Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland;
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| |
Collapse
|
12
|
Qiu X, Ye H, Li X, Li D, Jiang L, Liu R, Zhao Z, He D. IL-6/JAK2-dependent G6PD phosphorylation promotes nucleotide synthesis and supports tumor growth. Mol Metab 2023; 78:101836. [PMID: 37949355 PMCID: PMC10692918 DOI: 10.1016/j.molmet.2023.101836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/16/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023] Open
Abstract
OBJECTIVE Tumor cells hijack inflammatory mechanisms to promote their own growth. IL-6 is one of the major cytokines, and is frequently upregulated in tumors. The pentose phosphate pathway (PPP) generates the indispensable building blocks to produce various nucleotides. Here we aimed to determine whether and how PPP is timely tuned in response to IL-6 to support tumor growth. METHODS Protein expression was examined by immunoblot. Protein interaction was examined by immunoprecipitation. Tumor cell proliferation in in vitro culture was examined by BrdU assay and colony formation assay. Tumor cell proliferation in mouse xenograft model was examined by Ki-67 staining. RESULTS Here we show that the metabolic flux of PPP and enzymatic activity of glucose-6-phosphate dehydrogenase (G6PD) is rapidly induced under IL-6 treatment, without obvious changes in G6PD expression level. Mechanistically, Janus kinase 2 (JAK2) phosphorylates G6PD Y437 under IL-6 treatment, which accentuates G6PD enzymatic activity by promoting G6PD binding with its substrate G6P. Further, JAK2-dependent G6PD Y437 phosphorylation is required for IL-6-induced nucleotide biosynthesis and tumor cell proliferation, and is associated with the progression of oral squamous cell carcinoma. CONCLUSIONS Our findings report a new mechanism implicated in the crosstalk between tumor cells and inflammatory microenvironment, by which JAK2-dependent activation of G6PD governs nucleotide synthesis to support tumor cell proliferation, thereby highlighting its value as a potential anti-tumor target.
Collapse
Affiliation(s)
- Xuemei Qiu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Hongping Ye
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Xiaofei Li
- Department of Oncology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, 610057, PR China
| | - Dan Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Lu Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China.
| | - Rui Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China.
| | - Zhe Zhao
- Nuclear Stress Medicine Center, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, 610057, PR China.
| | - Dan He
- Department of Oncology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, 610057, PR China.
| |
Collapse
|
13
|
Jung J, Kim NH, Kwon M, Park J, Lim D, Kim Y, Gil W, Cheong YH, Park SA. The inhibitory effect of Gremlin-2 on adipogenesis suppresses breast cancer cell growth and metastasis. Breast Cancer Res 2023; 25:128. [PMID: 37880751 PMCID: PMC10599028 DOI: 10.1186/s13058-023-01732-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Gremlin-1 (GREM1) and Gremlin-2 (GREM2) are bone morphogenetic protein antagonists that play important roles in organogenesis, tissue differentiation, and tissue homeostasis. Although GREM1 has been reported to be involved in promoting various cancers, little has been reported about effects of GREM2 on cancer. Recently, it has been reported that GREM2 can inhibit adipogenesis in adipose-derived stromal/stem cells. However, as an inhibitor of adipogenesis, the role of GREM2 in cancer progression is not well understood yet. METHODS Pre-adipocyte 3T3-L1 cells overexpressing mock or Grem2 were established using a lentiviral transduction system and differentiated into adipocytes-mock and adipocytes-Grem2, respectively. To investigate the effect of adipocyte-Grem2 on breast cancer cells, we analyzed the proliferative and invasion abilities of spheroids using a 3D co-culture system of breast cancer cells and adipocytes or conditioned medium (CM) of adipocytes. An orthotopic breast cancer mouse model was used to examine the role of adipocytes-Grem2 in breast cancer progression. RESULTS Grem2 overexpression suppressed adipogenesis of 3T3-L1 cells. Proliferative and invasion abilities of spheroids formed by co-culturing MTV/TM-011 breast cancer cells and adipocytes-Grem2 were significantly reduced compared to those of spheroids formed by co-culturing MTV/TM-011 cells and adipocytes-mock. Compared to adipocytes-mock, adipocytes-Grem2 showed decreased mRNA expression of several adipokines, notably IL-6. The concentration of IL-6 in the CM of these cells was also decreased. Proliferative and invasive abilities of breast cancer cells reduced by adipocytes-Grem2 were restored by IL-6 treatment. Expression levels of vimentin, slug, and twist1 in breast cancer cells were decreased by treatment with CM of adipocytes-Grem2 but increased by IL-6 treatment. In orthotopic breast cancer mouse model, mice injected with both MTV/TM-011 cells and adipocytes-Grem2 showed smaller primary tumors and lower lung metastasis than controls. However, IL-6 administration increased both the size of primary tumor and the number of metastatic lung lesions, which were reduced by adipocytes-Grem2. CONCLUSIONS Our study suggests that GREM2 overexpression in adipocytes can inhibit adipogenesis, reduce the expression and secretion of several adipokines, including IL-6, and ultimately inhibit breast cancer progression.
Collapse
Affiliation(s)
- Jiwoo Jung
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan-si, 31538, Republic of Korea
| | - Na Hui Kim
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan-si, 31538, Republic of Korea
| | - Minji Kwon
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan-si, 31538, Republic of Korea
| | - Jayeon Park
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan-si, 31538, Republic of Korea
| | - Dayeon Lim
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan-si, 31538, Republic of Korea
| | - Youjin Kim
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan-si, 31538, Republic of Korea
| | - World Gil
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan-si, 31538, Republic of Korea
| | - Ye Hwang Cheong
- Drug Discovery Research Laboratories, Dong-A ST Co., Ltd., Yongin, 17073, Republic of Korea
| | - Sin-Aye Park
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan-si, 31538, Republic of Korea.
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan-si, 31538, Republic of Korea.
| |
Collapse
|
14
|
Somu P, Basavegowda N, Gomez LA, Jayaprakash HV, Puneetha GK, Yadav AK, Paul S, Baek KH. Crossroad between the Heat Shock Protein and Inflammation Pathway in Acquiring Drug Resistance: A Possible Target for Future Cancer Therapeutics. Biomedicines 2023; 11:2639. [PMID: 37893013 PMCID: PMC10604354 DOI: 10.3390/biomedicines11102639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
The development of multidrug resistance (MDR) against chemotherapeutic agents has become a major impediment in cancer therapy. Understanding the underlying mechanism behind MDR can guide future treatment for cancer with better therapeutic outcomes. Recent studies evidenced that crossroads interaction between the heat shock proteins (HSP) and inflammatory responses under the tumor microenvironment plays a pivotal role in modulating drug responsiveness and drug resistance through a complex cytological process. This review aims to investigate the interrelationship between inflammation and HSP in acquiring multiple drug resistance and investigate strategies to overcome the drug resistance to improve the efficacy of cancer treatment. HSP plays a dual regulatory effect as an immunosuppressive and immunostimulatory agent, involving the simultaneous blockade of multiple signaling pathways in acquiring MDR. For example, HSP27 shows biological effects on monocytes by causing IL10 and TNFα secretion and blocking monocyte differentiation to normal dendritic cells and tumor-associated macrophages to promote cancer progression and chemoresistance. Thus, the HSP function and immune-checkpoint release modalities provide a therapeutic target for a therapeutically beneficial approach for enhancing anti-tumor immune responses. The interconnection between inflammation and HSP, along with the tumor microenvironment in acquiring drug resistance, has become crucial for rationalizing the effect of HSP immunomodulatory activity with immune checkpoint blockade. This relationship can overcome drug resistance and assist in the development of novel combinatorial cancer immunotherapy in fighting cancer with decreasing mortality rates.
Collapse
Affiliation(s)
- Prathap Somu
- Department of Biotechnology and Chemical Engineering, School of Civil & Chemical Engineering, Manipal University Jaipur, Dehmi Kalan, Jaipur 303007, India;
| | - Nagaraj Basavegowda
- Department of Biotechnology, Yeungnam University, Gyeongsan 38451, Republic of Korea;
| | - Levin Anbu Gomez
- Department of Biotechnology, School of Agriculture and Bioscience, Karunya Institute of Technology and Sciences (Deemed-to-be University), Karunya Nagar, Coimbatore 641114, India;
| | | | | | - Akhilesh Kumar Yadav
- Department of Environmental Engineering and Management, Chaoyang University of Technology, Taichung 413310, Taiwan;
| | - Subhankar Paul
- Structural Biology and Nanomedicine Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, India
| | - Kwang-Hyun Baek
- Department of Biotechnology, Yeungnam University, Gyeongsan 38451, Republic of Korea;
| |
Collapse
|
15
|
Schaff DL, Fasse AJ, White PE, Vander Velde RJ, Shaffer SM. Clonal differences underlie variable responses to sequential and prolonged treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.24.534152. [PMID: 36993721 PMCID: PMC10055379 DOI: 10.1101/2023.03.24.534152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Cancer cells exhibit dramatic differences in gene expression at the single-cell level which can predict whether they become resistant to treatment. Treatment perpetuates this heterogeneity, resulting in a diversity of cell states among resistant clones. However, it remains unclear whether these differences lead to distinct responses when another treatment is applied or the same treatment is continued. In this study, we combined single-cell RNA-sequencing with barcoding to track resistant clones through prolonged and sequential treatments. We found that cells within the same clone have similar gene expression states after multiple rounds of treatment. Moreover, we demonstrated that individual clones have distinct and differing fates, including growth, survival, or death, when subjected to a second treatment or when the first treatment is continued. By identifying gene expression states that predict clone survival, this work provides a foundation for selecting optimal therapies that target the most aggressive resistant clones within a tumor.
Collapse
|
16
|
Interleukin-6 and Hypoxia Synergistically Promote EMT-Mediated Invasion in Epithelial Ovarian Cancer via the IL-6/STAT3/HIF-1 α Feedback Loop. Anal Cell Pathol (Amst) 2023; 2023:8334881. [PMID: 36814597 PMCID: PMC9940980 DOI: 10.1155/2023/8334881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/27/2022] [Accepted: 01/15/2023] [Indexed: 02/15/2023] Open
Abstract
Extensive peritoneal spread and capacity for distant metastasis account for the majority of mortality from epithelial ovarian cancer (EOC). Accumulating evidence shows that interleukin-6 (IL-6) promotes tumor invasion and migration in EOC, although the molecular mechanisms remain to be fully elucidated. Meanwhile, the hypoxic microenvironment has been recognized to cause metastasis by triggering epithelial-mesenchymal transition (EMT) in several types of cancers. Here, we studied the synergy between IL-6 and hypoxia in inducing EMT in two EOC cell lines, A2780 cells and SKOV3 cells. Exogenous recombination of IL-6 and autocrine production of IL-6 regulated by plasmids both induced EMT phenotype in EOC cells characterized by downregulated E-cadherin as well as upregulated expression of vimentin and EMT-related transcription factors. The combined effects of IL-6 and hypoxia were more significant than those of either one treatment on EMT. Suppression of hypoxia-inducible factor-1α (HIF-1α) before IL-6 treatment inhibited the EMT phenotype and invasion ability of EOC cells, indicating that HIF-1α occupies a key position in the regulatory pathway of EMT associated with IL-6. EMT score was found positively correlated with mRNA levels of IL-6, signal transducer and activator of transcription 3 (STAT3), and HIF-1α, respectively, in 489 ovarian samples from The Cancer Genome Atlas dataset. Next, blockade of the abovementioned molecules by chemical inhibitors reversed the alteration in the protein levels of EMT markers induced by either exogenous or endogenous IL-6. These findings indicate a positive feedback loop between IL-6 and HIF-1α, and induce and maintain EMT phenotype through STAT3 signaling, which might provide a novel rationale for prognostic prediction and therapeutic targets in EOC.
Collapse
|
17
|
Kampan NC, Kartikasari AER, Deceneux C, Madondo MT, McNally OM, Flanagan KL, Aziz NA, Stephens AN, Reynolds J, Quinn MA, Plebanski M. Combining TNFR2-Expressing Tregs and IL-6 as Superior Diagnostic Biomarkers for High-Grade Serous Ovarian Cancer Masses. Cancers (Basel) 2023; 15:667. [PMID: 36765633 PMCID: PMC9913655 DOI: 10.3390/cancers15030667] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/14/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
We hypothesised that the inclusion of immunosuppressive and inflammatory biomarkers in HGSOC patients would improve the sensitivity and specificity of the preoperative marker prediction of malignancy in patients with ovarian masses. We tested a panel of 29 soluble immune factors by multiplex bead immunoassay and 16 phenotypic T cell markers by flow cytometry in pre-treatment blood samples from 66 patients undergoing surgery for suspected ovarian cancer or ovarian cancer risk reduction. The potential diagnostic utility of all parameters was explored using Volcano plots, principal component analysis (PCA) and receiver operator characteristic (ROC) analysis. We also assessed the effect of culturing PBMCs from 20 healthy donors in the presence of malignant ascites fluid. The combination of TNFR2+ Tregs and IL-6 in the pre-treatment blood of patients with advanced HGSOC effectively discriminated patients with benign or malignant ovarian masses. In vitro culturing of the PBMCs of healthy donors in malignant ascites promoted an increase in TNFR2-expressing Tregs, which were decreased following blockade with IL-6 or STAT3 activity. Pre-treatment serum IL-6 and peripheral blood TNFR2+ Tregs may be potential clinical biomarkers that can discriminate patients with malignant compared to benign ovarian cancer masses, and the relationship between IL-6 and TNFR2+ Treg is likely to be mediated via the STAT3 signalling pathway.
Collapse
Affiliation(s)
- Nirmala Chandralega Kampan
- Department of Immunology & Pathology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
- Oncology Unit, Royal Women’s Hospital, 20 Flemington Road, Parkville, VIC 3052, Australia
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | | | - Cyril Deceneux
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| | - Mutsa Tatenda Madondo
- Department of Immunology & Pathology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
| | - Orla M. McNally
- Oncology Unit, Royal Women’s Hospital, 20 Flemington Road, Parkville, VIC 3052, Australia
- Department of Obstetrics and Gynaecology, Melbourne University, Parkville, VIC 3052, Australia
| | - Katie Louise Flanagan
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
- School of Health Sciences and School of Medicine, University of Tasmania, Hobart, TAS 7005, Australia
| | - Norhaslinda A. Aziz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Andrew N. Stephens
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3800, Australia
- Epworth Research Institute, Epworth Healthcare, Richmond, VIC 3121, Australia
| | - John Reynolds
- Biostatistics Consulting Platform, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
| | - Michael A. Quinn
- Oncology Unit, Royal Women’s Hospital, 20 Flemington Road, Parkville, VIC 3052, Australia
| | - Magdalena Plebanski
- Department of Immunology & Pathology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| |
Collapse
|
18
|
Zhang S, Yu X, Xie Y, Ye G, Guo J. tRNA derived fragments:A novel player in gene regulation and applications in cancer. Front Oncol 2023; 13:1063930. [PMID: 36761955 PMCID: PMC9904238 DOI: 10.3389/fonc.2023.1063930] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023] Open
Abstract
The heterogeneous species of tRNA-derived fragments (tRFs) with specific biological functions was recently identified. Distinct roles of tRFs in tumor development and viral infection, mediated through transcriptional and post-transcriptional regulation, has been demonstrated. In this review, we briefly summarize the current literatures on the classification of tRFs and the effects of tRNA modification on tRF biogenesis. Moreover, we highlight the tRF repertoire of biological roles such as gene silencing, and regulation of translation, cell apoptosis, and epigenetics. We also summarize the biological roles of various tRFs in cancer development and viral infection, their potential value as diagnostic and prognostic biomarkers for different types of cancers, and their potential use in cancer therapy.
Collapse
Affiliation(s)
- Shuangshuang Zhang
- Department of Gastroenterology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China,Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, School of Medicine, Ningbo University, Ningbo, China
| | - Xiuchong Yu
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, School of Medicine, Ningbo University, Ningbo, China
| | - Yaoyao Xie
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, School of Medicine, Ningbo University, Ningbo, China
| | - Guoliang Ye
- Department of Gastroenterology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China,Institute of Digestive Diseases, Ningbo University, Ningbo, China
| | - Junming Guo
- Department of Gastroenterology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China,Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, School of Medicine, Ningbo University, Ningbo, China,Institute of Digestive Diseases, Ningbo University, Ningbo, China,*Correspondence: Junming Guo,
| |
Collapse
|
19
|
Abstract
The RAS family of proteins is among the most frequently mutated genes in human malignancies. In ovarian cancer (OC), the most lethal gynecological malignancy, RAS, especially KRAS mutational status at codons 12, 13, and 61, ranges from 6-65% spanning different histo-types. Normally RAS regulates several signaling pathways involved in a myriad of cellular signaling cascades mediating numerous cellular processes like cell proliferation, differentiation, invasion, and death. Aberrant activation of RAS leads to uncontrolled induction of several downstream signaling pathways such as RAF-1/MAPK (mitogen-activated protein kinase), PI3K phosphoinositide-3 kinase (PI3K)/AKT, RalGEFs, Rac/Rho, BRAF (v-Raf murine sarcoma viral oncogene homolog B), MEK1 (mitogen-activated protein kinase kinase 1), ERK (extracellular signal-regulated kinase), PKB (protein kinase B) and PKC (protein kinase C) involved in cell proliferation as well as maintenance pathways thereby driving tumorigenesis and cancer cell propagation. KRAS mutation is also known to be a biomarker for poor outcome and chemoresistance in OC. As a malignancy with several histotypes showing varying histopathological characteristics, we focus on reviewing recent literature showcasing the involvement of oncogenic RAS in mediating carcinogenesis and chemoresistance in OC and its subtypes.
Collapse
Affiliation(s)
- Lubna Therachiyil
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, 2713, Qatar
| | - Anjana Anand
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
| | | | | | - Hesham M. Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, 2713, Qatar
| | - Shahab Uddin
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
| |
Collapse
|
20
|
Kumar N, Vyas A, Agnihotri SK, Chattopadhyay N, Sachdev M. Small secretory proteins of immune cells can modulate gynecological cancers. Semin Cancer Biol 2022; 86:513-531. [PMID: 35150864 DOI: 10.1016/j.semcancer.2022.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 01/27/2023]
Abstract
Small secretory proteins of immune cells are mostly Cytokines, which include chemokines, interleukins, interferons, lymphokines and tumor necrosis factors but not hormones or growth factors. These secretory proteins are the molecular messengers and primarily involved in autocrine, paracrine and endocrine signaling as immunomodulating agents. Hence, these proteins actually regulate the cells of immune system to communicate with one another to produce a synchronized, robust, still self-regulated response to a specific antigen. Chemokines are smaller secreted proteins that control overall immune cell movement and location; these chemokines are divided into 4 subgroups, namely, CXC, CC, CX3C and C according to the position of 4 conserved cysteine residues. Complete characterization of cytokines and chemokines can exploit their vast signaling networks to develop cancer treatments. These secretory proteins like IL-6, IL-10, IL-12, TNFα, CCL2, CXCL4 & CXCL8 are predominantly expressed in most of the gynecological cancers, which directly stimulate immune effector cells and stromal cells at the tumor site and augment tumor cell recognition by cytotoxic T-cells. Hence; these secretory proteins are the major regulators, which can actually modulate all kinds of gynecological cancers. Furthermore, advancements in adoptive T-cell treatment have relied on the use of multiple cytokines/chemokines to establish a highly regulated environment for anti-tumor T cell growth. A number of in vitro studies as well as animal models and clinical subjects have also shown that cytokines/chemokines have broad antitumor activity, which has been translated into a number of cancer therapy approaches. This review will focus on the foremost cytokines & chemokines involved in the majority of the gynecological malignancies and discuss their basic biology as well as clinical applications.
Collapse
Affiliation(s)
- Niranjan Kumar
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India
| | - Akanksha Vyas
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India
| | | | - Naibedya Chattopadhyay
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India.
| | - Monika Sachdev
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India.
| |
Collapse
|
21
|
Lopatina T, Sarcinella A, Brizzi MF. Tumour Derived Extracellular Vesicles: Challenging Target to Blunt Tumour Immune Evasion. Cancers (Basel) 2022; 14:cancers14164020. [PMID: 36011012 PMCID: PMC9406972 DOI: 10.3390/cancers14164020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Tumour onset and development occur because of specific immune support. The immune system, which is originally able to perceive and eliminate incipient cancer cells, becomes suppressed and hijacked by cancer. For these purposes, tumour cells use extracellular vesicles (TEVs). Specific molecular composition allows TEVs to reprogram immune cells towards tumour tolerance. Circulating TEVs move from their site of origin to other organs, preparing “a fertile soil” for metastasis formation. This implies that TEV molecular content can provide a valuable tool for cancer biomarker discovery and potential targets to reshape the immune system into tumour recognition and eradication. Abstract Control of the immune response is crucial for tumour onset and progression. Tumour cells handle the immune reaction by means of secreted factors and extracellular vesicles (EV). Tumour-derived extracellular vesicles (TEV) play key roles in immune reprogramming by delivering their cargo to different immune cells. Tumour-surrounding tissues also contribute to tumour immune editing and evasion, tumour progression, and drug resistance via locally released TEV. Moreover, the increase in circulating TEV has suggested their underpinning role in tumour dissemination. This review brings together data referring to TEV-driven immune regulation and antitumour immune suppression. Attention was also dedicated to TEV-mediated drug resistance.
Collapse
|
22
|
Guo Y, Wang M, Zou Y, Jin L, Zhao Z, Liu Q, Wang S, Li J. Mechanisms of chemotherapeutic resistance and the application of targeted nanoparticles for enhanced chemotherapy in colorectal cancer. J Nanobiotechnology 2022; 20:371. [PMID: 35953863 PMCID: PMC9367166 DOI: 10.1186/s12951-022-01586-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/04/2022] [Indexed: 11/10/2022] Open
Abstract
Colorectal cancer is considered one of the major malignancies that threaten the lives and health of people around the world. Patients with CRC are prone to post-operative local recurrence or metastasis, and some patients are advanced at the time of diagnosis and have no chance for complete surgical resection. These factors make chemotherapy an indispensable and important tool in treating CRC. However, the complex composition of the tumor microenvironment and the interaction of cellular and interstitial components constitute a tumor tissue with high cell density, dense extracellular matrix, and high osmotic pressure, inevitably preventing chemotherapeutic drugs from entering and acting on tumor cells. As a result, a novel drug carrier system with targeted nanoparticles has been applied to tumor therapy. It can change the physicochemical properties of drugs, facilitate the crossing of drug molecules through physiological and pathological tissue barriers, and increase the local concentration of nanomedicines at lesion sites. In addition to improving drug efficacy, targeted nanoparticles also reduce side effects, enabling safer and more effective disease diagnosis and treatment and improving bioavailability. In this review, we discuss the mechanisms by which infiltrating cells and other stromal components of the tumor microenvironment comprise barriers to chemotherapy in colorectal cancer. The research and application of targeted nanoparticles in CRC treatment are also classified.
Collapse
Affiliation(s)
- Yu Guo
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Min Wang
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Yongbo Zou
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Longhai Jin
- Department of Radiology, Jilin University Second Hospital, Changchun, 130000, China
| | - Zeyun Zhao
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Qi Liu
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Shuang Wang
- Department of the Dermatology, Jilin University Second Hospital, Changchun, 130000, China.
| | - Jiannan Li
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China.
| |
Collapse
|
23
|
Pre-diagnosis and post-diagnosis dietary patterns and survival in women with ovarian cancer. Br J Cancer 2022; 127:1097-1105. [DOI: 10.1038/s41416-022-01901-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 11/08/2022] Open
|
24
|
Piscazzi A, Condelli V, Crispo F, Coda ARD, Calice G, Bruno G, Venuto S, Tibullo D, Giordano G, Pietrafesa M, Liso A, Landriscina M. Differential and divergent activity of insulin-like growth factor binding protein 6 in platinum-sensitive versus platinum-resistant high-grade serous ovarian carcinoma cell lines. Oncol Lett 2022; 23:185. [PMID: 35527787 PMCID: PMC9073571 DOI: 10.3892/ol.2022.13305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/11/2022] [Indexed: 11/08/2022] Open
Abstract
Insulin-like growth factor binding protein 6 (IGFBP6) is a secreted protein with a controversial role in human malignancies, being downregulated in most types of human cancer, but upregulated in selected tumors. Ovarian cancer (OC) is a human malignancy characterized by IGFBP6 downregulation; however, the significance of its low expression during ovarian carcinogenesis is still poorly understood. In the present study, IGFBP6 expression and activation of its associated signaling pathway were evaluated in two matched OC cell lines derived from a high-grade serous OC before and after platinum resistance (PEA1 and PEA2 cells, respectively). A whole genome gene expression analysis was comparatively performed in both cell lines upon IGFBP6 stimulation using Illumina technology. IGFBP6 gene expression data from human OC cases were obtained from public datasets. Gene expression data from public datasets confirmed the downregulation of IGFBP6 in primary and metastatic OC tissues compared with in normal ovarian tissues. The comparative analysis of platinum-sensitive (PEA1) and platinum-resistant (PEA2) cell lines showed quantitative and qualitative differences in the activation of IGFBP6 signaling. Notably, IGFBP6 enhanced ERK1/2 phosphorylation only in PEA1 cells, and induced more evident and significant gene expression reprogramming in PEA1 cells compared with in PEA2 cells. Furthermore, the analysis of selected genes modulated by IGFBP6 (i.e., FOS, JUN, TNF, IL6, IL8 and EGR1) exhibited an inverse regulation in PEA1 versus PEA2 cells. In addition, selected hallmarks (TNFA_signaling_via_NFKB, TGF_beta_signaling, P53_pathway) and IL-6 signaling were positively regulated in PEA1 cells, whereas they were inhibited in PEA2 cells in response to IGFBP6. These data suggested that dysregulation of IGFBP6 signaling may serve a role in the progression of OC, and is likely associated with the development of platinum resistance.
Collapse
Affiliation(s)
- Annamaria Piscazzi
- Department of Medical and Surgical Sciences, University of Foggia, I-71122 Foggia, Italy
| | - Valentina Condelli
- Laboratory of Pre-Clinical and Translational Research, Scientific Institute for Research, Hospitalization and Healthcare-Referral Cancer Center of Basilicata (IRCCS-CROB), I-85028 Potenza, Italy
| | - Fabiana Crispo
- Laboratory of Pre-Clinical and Translational Research, Scientific Institute for Research, Hospitalization and Healthcare-Referral Cancer Center of Basilicata (IRCCS-CROB), I-85028 Potenza, Italy
| | - Anna Rita Daniela Coda
- Department of Medical and Surgical Sciences, University of Foggia, I-71122 Foggia, Italy
| | - Giovanni Calice
- Laboratory of Pre-Clinical and Translational Research, Scientific Institute for Research, Hospitalization and Healthcare-Referral Cancer Center of Basilicata (IRCCS-CROB), I-85028 Potenza, Italy
| | - Giuseppina Bruno
- Department of Medical and Surgical Sciences, University of Foggia, I-71122 Foggia, Italy
| | - Santina Venuto
- Department of Medical and Surgical Sciences, University of Foggia, I-71122 Foggia, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, I-95123 Catania, Italy
| | - Guido Giordano
- Department of Medical and Surgical Sciences, University of Foggia, I-71122 Foggia, Italy
| | - Michele Pietrafesa
- Laboratory of Pre-Clinical and Translational Research, Scientific Institute for Research, Hospitalization and Healthcare-Referral Cancer Center of Basilicata (IRCCS-CROB), I-85028 Potenza, Italy
| | - Arcangelo Liso
- Department of Medical and Surgical Sciences, University of Foggia, I-71122 Foggia, Italy
| | - Matteo Landriscina
- Department of Medical and Surgical Sciences, University of Foggia, I-71122 Foggia, Italy
- Laboratory of Pre-Clinical and Translational Research, Scientific Institute for Research, Hospitalization and Healthcare-Referral Cancer Center of Basilicata (IRCCS-CROB), I-85028 Potenza, Italy
| |
Collapse
|
25
|
Interleukin-6 Signaling in Triple Negative Breast Cancer Cells Elicits the Annexin A1/Formyl Peptide Receptor 1 Axis and Affects the Tumor Microenvironment. Cells 2022; 11:cells11101705. [PMID: 35626741 PMCID: PMC9139391 DOI: 10.3390/cells11101705] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 05/06/2022] [Accepted: 05/16/2022] [Indexed: 11/17/2022] Open
Abstract
Annexin A1 (AnxA1) is a pleiotropic protein that exerts essential roles in breast cancer (BC) growth and aggressiveness. In our previous work, we described the autocrine signaling of AnxA1 through formyl peptide receptor 1 (FPR1) in the triple-negative (TN) BC cell line, MDA-MB-231. Here, we aimed to describe the interaction between the AnxA1/FPR1 and the Interleukin-6 (IL-6) signaling pathways and their role in the tumor microenvironment (TME). First, we demonstrated that AnxA1 and IL-6 expression levels are correlated in BC tissue samples. In three TNBC cell lines, overexpression of both AnxA1 and IL-6 was also identified. Next, we inhibited FPR1, the IL-6 receptor and STAT3 in both MDA-MB-231 and MDA-MB-157 cells. The FPR1 inhibition led to increased levels of IL-6 and secreted AnxA1 in both cell lines. On the other side, inhibition of the IL-6 receptor or STAT3 led to the impairment of AnxA1 secretion, suggesting the essential role of the IL-6 signaling cascade in the activation of the AnxA1/FPR1 autocrine axis. Finally, we described the interaction between IL-6 and the AnxA1/FPR1 pathways and their role on the TME by analyzing the effect of supernatants derived from MDA-MB-231 and MDA-MB-157 cells under the inhibition of FPR1 or IL-6 signaling on fibroblast cell motility.
Collapse
|
26
|
Zhang R, Roque DM, Reader J, Lin J. Combined inhibition of IL‑6 and IL‑8 pathways suppresses ovarian cancer cell viability and migration and tumor growth. Int J Oncol 2022; 60:50. [PMID: 35315502 PMCID: PMC8973967 DOI: 10.3892/ijo.2022.5340] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 10/13/2021] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological cancer type in the United States. The success of current chemotherapies is limited by chemoresistance and side effects. Targeted therapy is a promising future direction for cancer therapy. In the present study, the efficacy of co‑targeting IL‑6 and IL‑8 in human ovarian cancer cells by bazedoxifene (Baze) + SCH527123 (SCH) treatment was examined. ELISA, cell viability, cell proliferation, cell migration, cell invasion, western blotting and peritoneal ovarian tumor mouse model analyses were performed to analyze the expression levels of IL‑6 and IL‑8, tumor growth, tumor migration and invasion, and the possible pathways of human ovarian cancer cell lines (SKOV3, CAOV3 and OVCAR3) and patient‑derived OV75 ovarian cancer cells. Each cell line was treated by monotherapy or combination therapy. The results demonstrated that IL‑6 and IL‑8 were secreted by human ovarian cancer cell lines. Compared with the DMSO control, the combination of IL‑6/glycoprotein 130 inhibitor Baze and IL‑8 inhibitor SCH synergistically inhibited cell viability in ovarian cancer cells. Baze + SCH also inhibited cell migration and invasion, suppressed ovarian tumor growth and inhibited STAT3 and AKT phosphorylation, as well as survivin expression. Therefore, co‑targeting the IL‑6 and IL‑8 signaling pathways may be an effective approach for ovarian cancer treatment.
Collapse
Affiliation(s)
- Ruijie Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Dana M Roque
- Division of Gynecologic Oncology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jocelyn Reader
- Division of Gynecologic Oncology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jiayuh Lin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
27
|
Extracellular Vesicle-Mediated IL-1 Signaling in Response to Doxorubicin Activates PD-L1 Expression in Osteosarcoma Models. Cells 2022; 11:cells11061042. [PMID: 35326493 PMCID: PMC8946890 DOI: 10.3390/cells11061042] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 02/04/2023] Open
Abstract
The expression of programmed cell death ligand 1 (PD-L1) in tumors is associated with tumor cell escape from T-cell cytotoxicity, and is considered a crucial effector in chemoresistance and tumor relapse. Although PD-L1 induction has been observed in patients after chemotherapy treatment, the mechanism by which the drug activates PD-L1 expression remains elusive. Here, we identified the extracellular vesicles (EVs) as a molecular mediator that determines the effect of doxorubicin on PD-L1 expression in osteosarcoma models. Mechanistically, doxorubicin dependently stimulates the release of extracellular vesicles, which mediate autocrine/paracrine signals in osteosarcoma cells. The recipient cells were stimulated by these EVs and acquired the ability to promote the expression of inflammatory cytokines interleukin (IL)-1β and IL-6. In response to doxorubicin, IL-1β, but not IL-6, allowed- osteosarcoma cells to promote the expression of PD-L1, and the elimination of IL-1β/IL-1 receptor signaling with IL-1 receptor antagonist reduced PD-L1 expression. Together, these findings provided insights into the role of EV release in response to chemotherapy that mediates PD-L1 expression via the IL-1 signaling pathway, and suggested that the combination of a drug targeting IL-1 or PD-L1 with chemotherapy could be an effective treatment option for osteosarcoma patients.
Collapse
|
28
|
Lee AH, Mejia Peña C, Dawson MR. Comparing the Secretomes of Chemorefractory and Chemoresistant Ovarian Cancer Cell Populations. Cancers (Basel) 2022; 14:1418. [PMID: 35326569 PMCID: PMC8946241 DOI: 10.3390/cancers14061418] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
High-grade serous ovarian cancer (HGSOC) constitutes the majority of all ovarian cancer cases and has staggering rates of both refractory and recurrent disease. While most patients respond to the initial treatment with paclitaxel and platinum-based drugs, up to 25% do not, and of the remaining that do, 75% experience disease recurrence within the subsequent two years. Intrinsic resistance in refractory cases is driven by environmental stressors like tumor hypoxia which alter the tumor microenvironment to promote cancer progression and resistance to anticancer drugs. Recurrent disease describes the acquisition of chemoresistance whereby cancer cells survive the initial exposure to chemotherapy and develop adaptations to enhance their chances of surviving subsequent treatments. Of the environmental stressors cancer cells endure, exposure to hypoxia has been identified as a potent trigger and priming agent for the development of chemoresistance. Both in the presence of the stress of hypoxia or the therapeutic stress of chemotherapy, cancer cells manage to cope and develop adaptations which prime populations to survive in future stress. One adaptation is the modification in the secretome. Chemoresistance is associated with translational reprogramming for increased protein synthesis, ribosome biogenesis, and vesicle trafficking. This leads to increased production of soluble proteins and extracellular vesicles (EVs) involved in autocrine and paracrine signaling processes. Numerous studies have demonstrated that these factors are largely altered between the secretomes of chemosensitive and chemoresistant patients. Such factors include cytokines, growth factors, EVs, and EV-encapsulated microRNAs (miRNAs), which serve to induce invasive molecular, biophysical, and chemoresistant phenotypes in neighboring normal and cancer cells. This review examines the modifications in the secretome of distinct chemoresistant ovarian cancer cell populations and specific secreted factors, which may serve as candidate biomarkers for aggressive and chemoresistant cancers.
Collapse
Affiliation(s)
- Amy H. Lee
- Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA;
| | - Carolina Mejia Peña
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA;
| | - Michelle R. Dawson
- Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA;
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA;
| |
Collapse
|
29
|
Ritch SJ, Telleria CM. The Transcoelomic Ecosystem and Epithelial Ovarian Cancer Dissemination. Front Endocrinol (Lausanne) 2022; 13:886533. [PMID: 35574025 PMCID: PMC9096207 DOI: 10.3389/fendo.2022.886533] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is considered the deadliest gynecological disease and is normally diagnosed at late stages, at which point metastasis has already occurred. Throughout disease progression, EOC will encounter various ecosystems and the communication between cancer cells and these microenvironments will promote the survival and dissemination of EOC. The primary tumor is thought to develop within the ovaries or the fallopian tubes, both of which provide a microenvironment with high risk of causing DNA damage and enhanced proliferation. EOC disseminates by direct extension from the primary tumors, as single cells or multicellular aggregates. Under the influence of cellular and non-cellular factors, EOC spheroids use the natural flow of peritoneal fluid to reach distant organs within the peritoneal cavity. These cells can then implant and seed distant organs or tissues, which develop rapidly into secondary tumor nodules. The peritoneal tissue and the omentum are two common sites of EOC metastasis, providing a microenvironment that supports EOC invasion and survival. Current treatment for EOC involves debulking surgery followed by platinum-taxane combination chemotherapy; however, most patients will relapse with a chemoresistant disease with tumors developed within the peritoneum. Therefore, understanding the role of the unique microenvironments that promote EOC transcoelomic dissemination is important in improving patient outcomes from this disease. In this review article, we address the process of ovarian cancer cellular fate at the site of its origin in the secretory cells of the fallopian tube or in the ovarian surface epithelial cells, their detachment process, how the cells survive in the peritoneal fluid avoiding cell death triggers, and how cancer- associated cells help them in the process. Finally, we report the mechanisms used by the ovarian cancer cells to adhere and migrate through the mesothelial monolayer lining the peritoneum. We also discuss the involvement of the transcoelomic ecosystem on the development of chemoresistance of EOC.
Collapse
Affiliation(s)
- Sabrina J. Ritch
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Carlos M. Telleria
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada
- *Correspondence: Carlos M. Telleria, ; orcid.org/0000-0003-1070-3538
| |
Collapse
|
30
|
Rasool M, Malik A, Waquar S, Ain QT, Rasool R, Asif M, Anfinan N, Haque A, Alam H, Ahmed S, Hamid Hamdard M. Assessment of clinical variables as predictive markers in the development and progression of colorectal cancer. Bioengineered 2021; 12:2288-2298. [PMID: 34096454 PMCID: PMC8806642 DOI: 10.1080/21655979.2021.1933680] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is graded as one of the most common cancer. It accounts for the second leading cause of cancer deaths worldwide. The present study intends to investigate the role and importance of different biochemical variables in the development of colorectal cancer.In this cross-sectional study we recruited ninety-one patients diagnosed with colorectal cancer and fifty-three age-sex matched controls from June 2017 to June 2018. Different variables i.e. SOD, GSH, CAT, MDA, TGF, VEGF, TNF, ILs, MMPs, etc., were estimated with the help of their respective methods. Our findings suggest a significant increase in the levels of different inflammatory and stress-related markers. The NFκB, TGF-β, VEGFβ, 8OHdG, IsoP-2α were significantly found to be increased in patients with colon cancer (0.945 ± 0.067 μg/ml, 18.59 ± 1.53 pg/ml, 99.35 ± 4.29 pg/ml, 21.26 ± 1.29 pg/ml, 102.25 ± 4.25 pg/ml) as compared to controls (0.124 ± 0.024 μg/ml, 8.26 ± 0.88 pg/ml, 49.58 ± 2.62 pg/ml, 0.93 ± 0.29 pg/ml, 19.65 ± 3.19 pg/ml). Notably, the levels of different antioxidants were shown to be significantly lower in patients of colon cancer. The present study concluded that excessive oxidative stress and lipid peroxidation result in a decrease in the antioxidative capacity of cells which may influence diverse signaling cascades including NF-KB, which results in DNA modification and gene transcription that ultimately involved in the progression of colon cancer.
Collapse
Affiliation(s)
- Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Arif Malik
- Institute of Molecular Biology and Biotechnology, the University of Lahore, Lahore, Pakistan
| | - Sulayman Waquar
- Institute of Molecular Biology and Biotechnology, the University of Lahore, Lahore, Pakistan
| | - Qura Tul Ain
- Institute of Molecular Biology and Biotechnology, the University of Lahore, Lahore, Pakistan
| | - Rabia Rasool
- Institute of Molecular Biology and Biotechnology, the University of Lahore, Lahore, Pakistan
| | - Muhammad Asif
- Department of Biotechnology, BUITEMS, Quetta, Pakistan
- ORIC, Buitems, Quetta, Pakistan
| | - Nisreen Anfinan
- Gynecology Oncology Unit, Obstetrics and Gynaecology Department, Faculty of Medicine, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Absarul Haque
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hina Alam
- Pakistan Institute of Medical Sciences, Islamabad, Pakistan
| | - Sagheer Ahmed
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University Islamabad
| | | |
Collapse
|
31
|
Abdel-Latif RT, Wadie W, Abdel-mottaleb Y, Abdallah DM, El-Maraghy NN, El-Abhar HS. Reposition of the anti-inflammatory drug diacerein in an in-vivo colorectal cancer model. Saudi Pharm J 2021; 30:72-90. [PMID: 35145347 PMCID: PMC8802128 DOI: 10.1016/j.jsps.2021.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023] Open
|
32
|
Zhang R, Wang H, Li E, Wu Y, Wen Y, Li C, Liao B, Ma Q. Quantitative phosphoproteomic analysis reveals chemoresistance-related proteins and signaling pathways induced by rhIL-6 in human osteosarcoma cells. Cancer Cell Int 2021; 21:581. [PMID: 34717622 PMCID: PMC8557500 DOI: 10.1186/s12935-021-02286-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background IL-6 plays a pivotal role in resistance to chemotherapeutics, including lobaplatin. However, the underlying mechanisms are still unclear. This study was to investigate the changes in phosphoproteins and their related signaling pathways in the process of IL-6-induced chemoresistance to lobaplain in osteosarcoma cells. Methods We performed a quantitative phosphoproteomic analysis of the response of SaOS-2 osteosarcoma cells to recombinant human IL-6 (rhIL-6) intervention prior to lobaplatin treatment. The cells were divided into the control group (Con), the lobaplatin group (Lob), and the rhIL-6-and-lobaplatin group (IL-6). Three biological replicates of each group were included. The differentially expressed phosphoproteins were subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. Netphos 3.1 was used for the prediction of kinases, and STRING was used for the visualization of protein–protein interactions. The conserved motifs surrounding the phosphorylated residues were analyzed using the motif-x algorithm. Western blot analysis was performed to verify the differential expression of p-FLNC, its predicted kinase and the related signaling pathway. The results of the bioinformatic analysis were validated by immunohistochemical staining of clinical specimens. Results In total, 3373 proteins and 12,183 peptides, including 3232 phosphorylated proteins and 11,358 phosphorylated peptides, were identified and quantified. Twenty-three significantly differentially expressed phosphoproteins were identified in the comparison between the IL-6 and Lob groups, and p-FLNC ranked second among these phosphoproteins. GO and KEGG analyses revealed the pivotal role of mitogen-activated protein kinase signaling in drug resistance induced by rhIL-6. Four motifs, namely, -SPxxK-, -RxxSP-, -SP-, and -SPK-, demonstrated higher expression in the IL-6 group than in the Lob group. The western blot analysis results verified the higher expression of p-FLNC, AKT1, and p-ERK and the lower expression of p-JNK in the IL-6 group than in the Con and Lob groups. The immunohistochemical staining results showed that p-FLNC, AKT1 and p-ERK1/2 were highly expressed in platinum-resistant clinical specimens but weakly expressed in platinum-sensitive specimens, and platinum-resistant osteosarcoma specimens demonstrated weak expression of p-JNK. Conclusions This phosphoproteomic study is the first to reveal the signature associated with rhIL-6 intervention before lobaplatin treatment in human osteosarcoma cells. p-FLNC, AKT1, and MAPK signaling contributes to resistance to lobaplatin in osteosarcoma SaOS-2 cells and may represent molecular targets to overcome osteosarcoma chemoresistance. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02286-z.
Collapse
Affiliation(s)
- Rui Zhang
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Huan Wang
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Erliang Li
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yonghong Wu
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yanhua Wen
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Chenyu Li
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Bo Liao
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| | - Qiong Ma
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
33
|
Szulc-Kielbik I, Kielbik M, Nowak M, Klink M. The implication of IL-6 in the invasiveness and chemoresistance of ovarian cancer cells. Systematic review of its potential role as a biomarker in ovarian cancer patients. Biochim Biophys Acta Rev Cancer 2021; 1876:188639. [PMID: 34695534 DOI: 10.1016/j.bbcan.2021.188639] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/23/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022]
Abstract
Interleukin 6 (IL-6) is a pleiotropic cytokine that is strongly implicated in the development and progression of ovarian cancer. The most recognized actions of IL-6 in ovarian cancer (OC) cells are the induction of cell proliferation and inhibition of cell apoptosis. Equally important is its ability to enhance the migratory and invasive potential of OC cells. Moreover, the increased expression and secretion of this cytokine positively correlates with OC cell chemoresistance. Elevated concentrations of IL-6 are observed in the serum and ascites of ovarian cancer patients. Thus, its level is discussed in the literature as a potential biomarker that can help to discriminate malignant and nonmalignant ovarian tumors and allow for the prediction of the chemotherapy response. The importance of IL-6 in ovarian cancer is proved by the fact that this cytokine is a potential target to anti-cancer therapy. This review is divided into two parts. The first summarizes the general biological activity of IL-6, and overviews its impact on OC cells, as well as discusses the current proposition of IL-6 inclusion in combination of anti-OC therapy. The second part is a systematic review of IL-6 as a possible biomarker in ovarian cancer patients.
Collapse
Affiliation(s)
- Izabela Szulc-Kielbik
- Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland.
| | - Michal Kielbik
- Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland.
| | - Marek Nowak
- Dept. of Operative Gynecology and Gynecologic Oncology, Polish Mother's Memorial Hospital - Research Institute, 281/289 Rzgowska Str., Lodz, Poland; Dept. of Operative and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland.
| | - Magdalena Klink
- Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland.
| |
Collapse
|
34
|
Yan X, Cao M, Wang Z, Wang S, Chen Q. C-reactive protein promotes tongue squamous cell carcinoma chemoresistance by inhibiting the activation of caspase-3/9 via the CD64/AKT/mTOR pathway. Hum Cell 2021; 34:1424-1433. [PMID: 34019244 DOI: 10.1007/s13577-021-00555-7] [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: 12/14/2020] [Accepted: 05/18/2021] [Indexed: 10/21/2022]
Abstract
Recent studies have shown that C-reactive protein (CRP) participates in multiple types of cancer development. Here, the aim of this study was to investigate the role of CRP in tongue squamous cell carcinoma (TSCC) chemoresistance. Immunohistochemical staining showed that CRP expression was upregulated in TSCC tissues from cisplatin-resistant patients compared with that in cisplatin-sensitive TSCC samples. The CRP expression level was positively correlated with that of the drug-resistant marker MDR1. Moreover, functional experiments showed that CRP increased cell viability and decreased cisplatin-induced apoptosis. CRP also increased the expression levels of MDR1 and Bcl-2 and decreased the expression level of Bax. Furthermore, CRP decreased the activity of caspase-3. Mechanistically, CRP could bind to Fcγ receptor I (FcγRI, also known as CD64) and activate the AKT/mTOR pathway to inhibit the activation of caspase-3/9, as shown by co-immunoprecipitation (Co-IP) assay and western blotting assays. In addition, CRP promoted tumour growth and decreased cleaved caspase-3/9 expression in BALB/c nude mice. Taken together, our findings indicate that CRP promotes TSCC chemoresistance by inhibiting the activation of caspase-3/9 via the FcγRI/AKT/mTOR pathway. Thus, CRP could potentially be considered as a therapeutic target for reducing TSCC chemoresistance.
Collapse
Affiliation(s)
- Xiaodong Yan
- Department of Stomatology, Zibo Central Hospital, Zibo, 255036, China
| | - Meng Cao
- Department of Stomatology, Zibo Central Hospital, Zibo, 255036, China
| | - Zhigang Wang
- Department of Stomatology, Zibo Central Hospital, Zibo, 255036, China
| | - Shenglin Wang
- Department of Stomatology, Zibo Central Hospital, Zibo, 255036, China
| | - Qinchao Chen
- Department of Stomatology, Zibo Central Hospital, Zibo, 255036, China.
| |
Collapse
|
35
|
Morales-Guadarrama G, García-Becerra R, Méndez-Pérez EA, García-Quiroz J, Avila E, Díaz L. Vasculogenic Mimicry in Breast Cancer: Clinical Relevance and Drivers. Cells 2021; 10:cells10071758. [PMID: 34359928 PMCID: PMC8304745 DOI: 10.3390/cells10071758] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/24/2022] Open
Abstract
In solid tumors, vasculogenic mimicry (VM) is the formation of vascular structures by cancer cells, allowing to generate a channel-network able to transport blood and tumor cells. While angiogenesis is undertaken by endothelial cells, VM is assumed by cancer cells. Besides the participation of VM in tumor neovascularization, the clinical relevance of this process resides in its ability to favor metastasis and to drive resistance to antiangiogenic therapy. VM occurs in many tumor types, including breast cancer, where it has been associated with a more malignant phenotype, such as triple-negative and HER2-positive tumors. The latter may be explained by known drivers of VM, like hypoxia, TGFB, TWIST1, EPHA2, VEGF, matrix metalloproteinases, and other tumor microenvironment-derived factors, which altogether induce the transformation of tumor cells to a mesenchymal phenotype with a high expression rate of stemness markers. This review analyzes the current literature in the field, including the participation of some microRNAs and long noncoding RNAs in VM-regulation and tumorigenesis of breast cancer. Considering the clinical relevance of VM and its association with the tumor phenotype and clinicopathological parameters, further studies are granted to target VM in the clinic.
Collapse
Affiliation(s)
- Gabriela Morales-Guadarrama
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Rocío García-Becerra
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Edgar Armando Méndez-Pérez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Janice García-Quiroz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Euclides Avila
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
- Correspondence: ; Tel.: +52-(55)-5487-0900
| |
Collapse
|
36
|
Torii C, Maishi N, Kawamoto T, Morimoto M, Akiyama K, Yoshioka Y, Minami T, Tsumita T, Alam MT, Ochiya T, Hida Y, Hida K. miRNA-1246 in extracellular vesicles secreted from metastatic tumor induces drug resistance in tumor endothelial cells. Sci Rep 2021; 11:13502. [PMID: 34226586 PMCID: PMC8257582 DOI: 10.1038/s41598-021-92879-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
Tumor endothelial cells (TECs) reportedly exhibit altered phenotypes. We have demonstrated that TECs acquire drug resistance with the upregulation of P-glycoprotein (P-gp, ABCB1), contrary to traditional assumptions. Furthermore, P-gp expression was higher in TECs of highly metastatic tumors than in those of low metastatic tumors. However, the detailed mechanism of differential P-gp expression in TECs remains unclear. miRNA was identified in highly metastatic tumor extracellular vesicles (EVs) and the roles of miRNA in endothelial cell resistance were analyzed in vitro and in vivo. In the present study, we found that treatment of highly metastatic tumor-conditioned medium induced resistance to 5-fluorouracil (5-FU) with interleukin-6 (IL-6) upregulation in endothelial cells (ECs). Among the soluble factors secreted from highly metastatic tumors, we focused on EVs and determined that miR-1246 was contained at a higher level in highly metastatic tumor EVs than in low metastatic tumor EVs. Furthermore, miR-1246 was transported via the EVs into ECs and induced IL-6 expression. Upregulated IL-6 induced resistance to 5-FU with STAT3 and Akt activation in ECs in an autocrine manner. These results suggested that highly metastatic tumors induce drug resistance in ECs by transporting miR-1246 through EVs.
Collapse
Affiliation(s)
- Chisaho Torii
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan
- Department of Oral and Maxillofacial Surgery, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan
| | - Nako Maishi
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan
| | - Taisuke Kawamoto
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan
| | - Masahiro Morimoto
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan
- Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan
| | - Kosuke Akiyama
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan
| | - Yusuke Yoshioka
- Institute of Medical Science, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Takashi Minami
- Division of Molecular and Vascular Biology, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Takuya Tsumita
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan
| | - Mohammad Towfik Alam
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan
| | - Takahiro Ochiya
- Institute of Medical Science, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Faculty of Medicine, Sapporo, 060-8638, Japan
| | - Kyoko Hida
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan.
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan.
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, 060-8586, Japan.
| |
Collapse
|
37
|
Li X, Liu Y, Zheng S, Zhang T, Wu J, Sun Y, Zhang J, Liu G. Role of exosomes in the immune microenvironment of ovarian cancer. Oncol Lett 2021; 21:377. [PMID: 33777201 PMCID: PMC7988709 DOI: 10.3892/ol.2021.12638] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are excretory vesicles that can deliver a variety of bioactive cargo molecules to the extracellular environment. Accumulating evidence demonstrates exosome participation in intercellular communication, immune response, inflammatory response and they even play an essential role in affecting the tumor immune microenvironment. The role of exosomes in the immune microenvironment of ovarian cancer is mainly divided into suppression and stimulation. On one hand exosomes can stimulate the innate and adaptive immune systems by activating dendritic cells (DCs), natural killer cells and T cells, allowing these immune cells exert an antitumorigenic effect. On the other hand, ovarian cancer-derived exosomes initiate cross-talk with immunosuppressive effector cells, which subsequently cause immune evasion; one of the hallmarks of cancer. Exosomes induce the polarization of macrophages in M2 phenotype and induce apoptosis of lymphocytes and DCs. Exosomes further activate additional immunosuppressive effector cells (myeloid-derived suppressor cells and regulatory T cells) that induce fibroblasts to differentiate into cancer-associated fibroblasts. Exosomes also induce the tumorigenicity of mesenchymal stem cells to exert additional immune suppression. Furthermore, besides mediating the intercellular communication, exosomes carry microRNAs (miRNAs), proteins and lipids to the tumor microenvironment, which collectively promotes ovarian cancer cells to proliferate, invade and tumors to metastasize. Studying proteins, lipids and miRNAs carried by exosomes could potentially be used as an early diagnostic marker of ovarian cancer for designing treatment strategies.
Collapse
Affiliation(s)
- Xiao Li
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yang Liu
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shuangshuang Zheng
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Tianyu Zhang
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jing Wu
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yue Sun
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jingzi Zhang
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Guoyan Liu
- Department of Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
38
|
Beyranvand Nejad E, Labrie C, van Elsas MJ, Kleinovink JW, Mittrücker HW, Franken KLMC, Heink S, Korn T, Arens R, van Hall T, van der Burg SH. IL-6 signaling in macrophages is required for immunotherapy-driven regression of tumors. J Immunother Cancer 2021; 9:e002460. [PMID: 33879600 PMCID: PMC8061866 DOI: 10.1136/jitc-2021-002460] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND High serum interleukin (IL-6) levels may cause resistance to immunotherapy by modulation of myeloid cells in the tumor microenvironment. IL-6 signaling blockade is tested in cancer, but as this inflammatory cytokine has pleiotropic effects, this treatment is not always effective. METHODS IL-6 and IL-6R blockade was applied in an IL-6-mediated immunotherapy-resistant TC-1 tumor model (TC-1.IL-6) and immunotherapy-sensitive TC-1. CONTROL Effects on therapeutic vaccination-induced tumor regression, recurrence and survival as well on T cells and myeloid cells in the tumor microenvironment were studied. The effects of IL-6 signaling in macrophages under therapy conditions were studied in Il6rafl/fl×LysMcre+ mice. RESULTS Our therapeutic vaccination protocol elicits a strong tumor-specific CD8+ T-cell response, leading to enhanced intratumoral T-cell infiltration and recruitment of tumoricidal macrophages. Blockade of IL-6 signaling exacerbated tumor outgrowth, reflected by fewer complete regressions and more recurrences after therapeutic vaccination, especially in TC-1.IL-6 tumor-bearing mice. Early IL-6 signaling blockade partly inhibited the development of the vaccine-induced CD8+ T-cell response. However, the main mechanism was the malfunction of macrophages during therapy-induced tumor regression. Therapy efficacy was impaired in Il6rafl/fl×LysMcre+ but not cre-negative control mice, while no differences in the vaccine-induced CD8+ T-cell response were found between these mice. IL-6 signaling blockade resulted in decreased expression of suppressor of cytokine signaling 3, essential for effective M1-type function in macrophages, and increased expression of the phagocytic checkpoint molecule signal-regulatory protein alpha by macrophages. CONCLUSION IL-6 signaling is critical for macrophage function under circumstances of immunotherapy-induced tumor tissue destruction, in line with the acute inflammatory functions of IL-6 signaling described in infections.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Cytotoxicity, Immunologic/drug effects
- Female
- Injections, Subcutaneous
- Interleukin-6/metabolism
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasms/drug therapy
- Neoplasms/immunology
- Neoplasms/metabolism
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Papillomavirus E7 Proteins/administration & dosage
- Papillomavirus E7 Proteins/immunology
- Phenotype
- Receptors, Interleukin-6/genetics
- Receptors, Interleukin-6/metabolism
- Signal Transduction
- Tumor Burden/drug effects
- Tumor-Associated Macrophages/drug effects
- Tumor-Associated Macrophages/immunology
- Tumor-Associated Macrophages/metabolism
- Mice
Collapse
Affiliation(s)
- Elham Beyranvand Nejad
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Camilla Labrie
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Marit J van Elsas
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Jan Willem Kleinovink
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Hans-Willi Mittrücker
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kees L M C Franken
- Department of Immunology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Sylvia Heink
- Experimental Neuroimmunology, Technische Universität München, Munchen, Bayern, Germany
| | - Thomas Korn
- Experimental Neuroimmunology, Technische Universität München, Munchen, Bayern, Germany
| | - Ramon Arens
- Department of Immunology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Thorbald van Hall
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Sjoerd H van der Burg
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| |
Collapse
|
39
|
Le Naour A, Prat M, Thibault B, Mével R, Lemaitre L, Leray H, Joubert MV, Coulson K, Golzio M, Lefevre L, Mery E, Martinez A, Ferron G, Delord JP, Coste A, Couderc B. Tumor cells educate mesenchymal stromal cells to release chemoprotective and immunomodulatory factors. J Mol Cell Biol 2021; 12:202-215. [PMID: 31504643 PMCID: PMC7181721 DOI: 10.1093/jmcb/mjz090] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 06/05/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
Factors released by surrounding cells such as cancer-associated mesenchymal stromal cells (CA-MSCs) are involved in tumor progression and chemoresistance. In this study, we characterize the mechanisms by which naïve mesenchymal stromal cells (MSCs) can acquire a CA-MSCs phenotype. Ovarian tumor cells trigger the transformation of MSCs to CA-MSCs by expressing pro-tumoral genes implicated in the chemoresistance of cancer cells, resulting in the secretion of high levels of CXC chemokine receptors 1 and 2 (CXCR1/2) ligands such as chemokine (C-X-C motif) ligand 1 (CXCL1), CXCL2, and interleukin 8 (IL-8). CXCR1/2 ligands can also inhibit the immune response against ovarian tumor cells. Indeed, through their released factors, CA-MSCs promote the differentiation of monocytes towards M2 macrophages, which favors tumor progression. When CXCR1/2 receptors are inhibited, these CA-MSC-activated macrophages lose their M2 properties and acquire an anti-tumoral phenotype. Both ex vivo and in vivo, we used a CXCR1/2 inhibitor to sensitize ovarian tumor cells to carboplatin and circumvent the pro-tumoral effects of CA-MSCs. Since high concentrations of CXCR1/2 ligands in patients’ blood are associated with chemoresistance, CXCR1/2 inhibition could be a potential therapeutic strategy to revert carboplatin resistance.
Collapse
Affiliation(s)
- Augustin Le Naour
- Institut Claudius Regaud -IUCT Oncopole, Université de Toulouse, Toulouse, France.,INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France
| | - Mélissa Prat
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, Toulouse, France
| | - Benoît Thibault
- Institut Claudius Regaud -IUCT Oncopole, Université de Toulouse, Toulouse, France.,INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France
| | - Renaud Mével
- Institut Claudius Regaud -IUCT Oncopole, Université de Toulouse, Toulouse, France.,INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France
| | - Léa Lemaitre
- Institut Claudius Regaud -IUCT Oncopole, Université de Toulouse, Toulouse, France.,INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France
| | - Hélène Leray
- Institut Claudius Regaud -IUCT Oncopole, Université de Toulouse, Toulouse, France.,INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France
| | - Marie-Véronique Joubert
- Institut Claudius Regaud -IUCT Oncopole, Université de Toulouse, Toulouse, France.,INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France
| | - Kimberley Coulson
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, Toulouse, France
| | - Muriel Golzio
- UMR CNRS 5089, Institut de Pharmacologie et de Biologie Structurale (IPBS), Toulouse, France
| | - Lise Lefevre
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, Toulouse, France
| | - Eliane Mery
- Institut Claudius Regaud -IUCT Oncopole, Université de Toulouse, Toulouse, France
| | - Alejandra Martinez
- Institut Claudius Regaud -IUCT Oncopole, Université de Toulouse, Toulouse, France
| | - Gwénaël Ferron
- Institut Claudius Regaud -IUCT Oncopole, Université de Toulouse, Toulouse, France
| | - Jean-Pierre Delord
- Institut Claudius Regaud -IUCT Oncopole, Université de Toulouse, Toulouse, France.,INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France
| | - Agnès Coste
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, Toulouse, France
| | - Bettina Couderc
- Institut Claudius Regaud -IUCT Oncopole, Université de Toulouse, Toulouse, France.,INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France
| |
Collapse
|
40
|
Wang H, Li B, Yan K, Wu Y, Wen Y, Liu Y, Fan P, Ma Q. Protein and Signaling Pathway Responses to rhIL-6 Intervention Before Lobaplatin Treatment in Osteosarcoma Cells. Front Oncol 2021; 11:602712. [PMID: 33791202 PMCID: PMC8006349 DOI: 10.3389/fonc.2021.602712] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/16/2021] [Indexed: 01/28/2023] Open
Abstract
Lobaplatin is a third-generation platinum-based antineoplastic agent and is widely used for osteosarcoma treatment before and after tumor removal. However, treatment failure often results from lobaplatin drug resistance. In our study, we found that SaOS-2 and SOSP-9607 osteosarcoma cells became less sensitive to lobaplatin after treatment with exogenous interleukin (IL)-6. Quantitative proteomic analysis was performed to elucidate the underlying mechanism in SaOS-2 osteosarcoma cells. Cells were divided into a control group (CG), a lobaplatin treatment group (LG), a recombinant human IL-6 (rhIL-6), and a lobaplatin treatment group (rhILG). We performed three biological replicates in each group to compare the differential protein expression between groups using a tandem mass tag (TMT) labeling technology based on liquid chromatography-tandem mass spectrometry (LC-MS/MS). A total of 1,313 proteins with significant differential expression was identified and quantified. The general characteristics of the significantly enriched proteins were identified by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, and protein-protein interaction (PPI) analysis was conducted using IntAct and STRING. In total, 31 proteins were further verified by parallel reaction monitoring (PRM), among which ras GTPase-activating protein-binding protein 1 (G3BP1), fragile X mental retardation syndrome-related protein 1 (hFXR1p), and far upstream element-binding protein 1 (FUBP1) were significantly differentially expressed. Immunohistochemistry results showed that these three proteins are highly expressed in specimens from platinum-resistant osteosarcoma patients, while the proteins are negatively or weakly expressed in specimens from platinum-sensitive osteosarcoma patients. The immunofluorescence staining results were in accord with the immunohistochemistry staining results. siRNA knockdown of FUBP1 showed a strikingly decreased IC50 value for lobaplatin in FUBP1-silenced cells, which verified the role of FUBP1 in the drug susceptibility of osteosarcoma and the potential therapeutic value for increasing the sensitivity to lobaplatin. This is the first proteomic study on a rhIL-6 intervention before lobaplatin treatment in osteosarcoma cells.
Collapse
Affiliation(s)
- Huan Wang
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Bin Li
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Kang Yan
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yonghong Wu
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yanhua Wen
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yunyan Liu
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Pei Fan
- Department of Orthopedics, The Second Affiliated Hospital of Wenzhou Medical University, Yuying Children's Hospital, Wenzhou, China
| | - Qiong Ma
- Orthopedic Oncology Institute, Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
41
|
Tendulkar S, Dodamani S. Chemoresistance in Ovarian Cancer: Prospects for New Drugs. Anticancer Agents Med Chem 2021; 21:668-678. [PMID: 32900355 DOI: 10.2174/1871520620666200908104835] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/04/2020] [Accepted: 08/08/2020] [Indexed: 12/09/2022]
Abstract
This review focuses on the conventional treatment, signaling pathways and various reasons for drug resistance with an understanding of novel methods that can lead to effective therapies. Ovarian cancer is amongst the most common gynecological and lethal cancers in women affecting different age groups (20-60). The survival rate is limited to 5 years due to diagnosis in subsequent stages with a reoccurrence of tumor and resistance to chemotherapeutic therapy. The recent clinical trials use the combinatorial treatment of carboplatin and paclitaxel on ovarian cancer after the cytoreduction of the tumor. Predominantly, patients are responsive initially to therapy and later develop metastases due to drug resistance. Chemotherapy also leads to drug resistance causing enormous variations at the cellular level. Multifaceted mechanisms like drug resistance are associated with a number of genes and signaling pathways that process the proliferation of cells. Reasons for resistance include epithelial-mesenchyme, DNA repair activation, autophagy, drug efflux, pathway activation, and so on. Determining the routes on the molecular mechanism that target chemoresistance pathways are necessary for controlling the treatment and understanding efficient drug targets can open light on improving therapeutic outcomes. The most common drug used for ovarian cancer is Cisplatin that activates various chemoresistance pathways, ultimately causing drug resistance. There have been substantial improvements in understanding the mechanisms of cisplatin resistance or chemo sensitizing cisplatin for effective treatment. Therefore, using therapies that involve a combination of phytochemical or novel drug delivery system would be a novel treatment for cancer. Phytochemicals are plant-derived compounds that exhibit anti-cancer, anti-oxidative, anti-inflammatory properties and reduce side effects exerted by chemotherapeutics.
Collapse
Affiliation(s)
- Shivani Tendulkar
- Dr. Prabhakar Kore Basic Science Research Center, KLE Academy of Higher Education and Research, Nehru Nagar, Belagavi- 590010, Karnataka, India
| | - Suneel Dodamani
- Dr. Prabhakar Kore Basic Science Research Center, KLE Academy of Higher Education and Research, Nehru Nagar, Belagavi- 590010, Karnataka, India
| |
Collapse
|
42
|
Xu S, Yu C, Ma X, Li Y, Shen Y, Chen Y, Huang S, Zhang T, Deng W, Wang Y. IL-6 promotes nuclear translocation of HIF-1α to aggravate chemoresistance of ovarian cancer cells. Eur J Pharmacol 2021; 894:173817. [PMID: 33345849 DOI: 10.1016/j.ejphar.2020.173817] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/28/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022]
Abstract
The inflammatory milieu in tumor modulates the resistance to the conventional antitumoral therapies. Interleukin-6 (IL-6), a pleiotropic pro-inflammatory cytokine and a crucial mediator of tumor development, has been targeted as a therapeutic strategy to overcome chemoresistance in the treatment of tumors. The protein levels and nuclear translocation of HIFs (hypoxia-inducible factors), such as HIF-1α, are linked to the drug resistance of tumor cells. However, whether IL-6 promotes the nuclear translocation of HIF-1α and the related mechanism remain to be investigated. We applied two ovarian cancer (OvCa) cell lines, A2780 cells and SKOV3 cells for the in vivo and in vitro studies. We found that IL-6 up-regulates the HIF-1α expression via the signal transducer and activator of transcription 3 (STAT3) signaling under hypoxia in either endogenous or exogenous way, and then we proved that IL-6 enhances the transcriptional activity of HIF-1α via the STAT3 signaling. Further mechanism research revealed that IL-6 promotes the nuclear translocation of HIF-1α through the STAT3 signaling under hypoxia. Proliferation assay and apoptosis assay were applied and proved that IL-6 enhances the chemoresistance of OvCa cells against cisplatin through the upregulation of HIF-1α via the STAT3 signaling in vitro. The In vivo studies confirmed the effect of IL-6 in increasing the chemoresistance of OvCa cells against cisplatin through the IL-6/STAT3/HIF-1α loop in the animal models. Our data elucidates the explicit mechanism of IL-6/STAT3/HIF-1α loop in OvCa and also provides new insights into the development of different approaches for the inflammation-induced and hypoxia-induced resistance in tumor therapies.
Collapse
Affiliation(s)
- Shiwen Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Niusai Biotechnology Co., Ltd, Tianjin, 300381, China
| | - Chunyan Yu
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaoxia Ma
- Tianjin Medical University General Hospital Airport Hospital, Tianjin, 300308, China
| | - Yan Li
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Yangyang Shen
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China
| | - Yan Chen
- Ningbo Hangzhou Bay Hospital, Ningbo, Zhejiang Province, 315336, China
| | - Suhui Huang
- Department of Pathogenic Biology and Immunology, Logistics University of Chinese People's Armed Police Forces, Tianjin, 300309, China
| | - Tongshuo Zhang
- Department of Pathogenic Biology and Immunology, Logistics University of Chinese People's Armed Police Forces, Tianjin, 300309, China
| | - Weimin Deng
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, Tianjin, 300070, China.
| | - Yue Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
43
|
Cummings M, Freer C, Orsi NM. Targeting the tumour microenvironment in platinum-resistant ovarian cancer. Semin Cancer Biol 2021; 77:3-28. [PMID: 33607246 DOI: 10.1016/j.semcancer.2021.02.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/09/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Ovarian cancer typically presents at an advanced stage, and although the majority of cases initially respond well to platinum-based therapies, chemoresistance almost always occurs leading to a poor long-term prognosis. While various cellular autonomous mechanisms contribute to intrinsic or acquired platinum resistance, the tumour microenvironment (TME) plays a central role in resistance to therapy and disease progression by providing cancer stem cell niches, promoting tumour cell metabolic reprogramming, reducing chemotherapy drug perfusion and promoting an immunosuppressive environment. As such, the TME is an attractive therapeutic target which has been the focus of intense research in recent years. This review provides an overview of the unique ovarian cancer TME and its role in disease progression and therapy resistance, highlighting some of the latest preclinical and clinical data on TME-targeted therapies. In particular, it focuses on strategies targeting cancer-associated fibroblasts, tumour-associated macrophages, cancer stem cells and cancer cell metabolic vulnerabilities.
Collapse
Affiliation(s)
- M Cummings
- Leeds Institute of Medical Research at St James's, St James's University Hospital, Beckett Street, Leeds, LS9 7TF, United Kingdom
| | - C Freer
- Leeds Institute of Medical Research at St James's, St James's University Hospital, Beckett Street, Leeds, LS9 7TF, United Kingdom
| | - N M Orsi
- Leeds Institute of Medical Research at St James's, St James's University Hospital, Beckett Street, Leeds, LS9 7TF, United Kingdom; St James's Institute of Oncology, Bexley Wing, Beckett Street, Leeds, LS9 7TF, United Kingdom.
| |
Collapse
|
44
|
Huang SL, Chang TC, Chao CCK, Sun NK. TLR4/IL-6/IRF1 signaling regulates androgen receptor expression: A potential therapeutic target to overcome taxol resistance in ovarian cancer. Biochem Pharmacol 2021; 186:114456. [PMID: 33556340 DOI: 10.1016/j.bcp.2021.114456] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/24/2021] [Accepted: 01/28/2021] [Indexed: 10/22/2022]
Abstract
Ovarian cancer is poorly treatable due, at least in part, to induced drug resistance to taxol- and cisplatin-based chemotherapy. Recent studies showed that ectopic overexpression of toll-like receptor 4 (TLR4) in ovarian cancer cells leads to upregulation of the androgen receptor (AR) and transactivation of taxol resistance genes, thereby causing chemoresistance. In the present study, we examined the signaling pathways involving TLR4 and interleukin 6 (IL-6) that enhance AR expression. Based on transcriptomic analysis, we show that IL-6 functions as a hub gene among the upregulated genes in taxol-treated TLR4-overexpressing ovarian cancer cells. Both the TLR4 activator taxol and IL-6 can induce AKT phosphorylation, whereas TLR4 knockdown or inhibition of the IL-6 signal transducer GP130 abrogates AKT activation. Furthermore, expression of AR and IL-6 is downregulated in TLR4-knockdown, taxol-resistant cells. In addition, TLR4 knockdown inhibits GP130 and IL-6 receptor alpha (IL6Rα) activities, indicating that TLR4 plays a critical role in IL-6 signaling. On the other hand, nuclear translocation of AR is induced by IL-6 treatment, whereas knockdown of endogenous IL-6 reduces AR and TLR4 expression in taxol-resistant ovarian cancer cells. These results indicate that TLR4 and IL-6 play a crucial role in AR gene regulation and function. We also identify interferon regulatory factor 1 (IRF1) as a downstream target of IL-6 signaling and as a regulator of AR expression. Moreover, analysis of clinical samples indicates that high IL-6 expression correlates with poor progression-free survival in ovarian cancer patients treated with taxol. Overall, our findings indicate that the TLR4/IL-6/IRF1 signaling axis represents a potential therapeutic target to overcome AR-based taxol resistance in ovarian cancer.
Collapse
Affiliation(s)
- Shang-Lang Huang
- Division of Biomedical Sciences, Chang Gung University of Science and Technology, Taoyuan, Taiwan, ROC; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan, ROC.
| | - Ting-Chang Chang
- Department of Obstetrics and Gynaecology, Chang Gung Memorial Hospital Linkou Medical Centre, Taoyuan, Taiwan, ROC.
| | - Chuck C K Chao
- Department of Obstetrics and Gynaecology, Chang Gung Memorial Hospital Linkou Medical Centre, Taoyuan, Taiwan, ROC; Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC.
| | - Nian-Kang Sun
- Division of Biomedical Sciences, Chang Gung University of Science and Technology, Taoyuan, Taiwan, ROC; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan, ROC; Department of Obstetrics and Gynaecology, Chang Gung Memorial Hospital Linkou Medical Centre, Taoyuan, Taiwan, ROC.
| |
Collapse
|
45
|
Wang Z, Li F, Wei M, Zhang S, Wang T. Circadian Clock Protein PERIOD2 Suppresses the PI3K/Akt Pathway and Promotes Cisplatin Sensitivity in Ovarian Cancer. Cancer Manag Res 2020; 12:11897-11908. [PMID: 33244267 PMCID: PMC7683831 DOI: 10.2147/cmar.s278903] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/17/2020] [Indexed: 02/06/2023] Open
Abstract
Background The mortality rate of ovarian cancer is the highest among gynecological tumors. The two factors leading to high mortality of ovarian cancer are late clinical stage and chemotherapy resistance. It is very important to reverse or intervene chemotherapy resistance. Abnormal circadian rhythm is related to the occurrence of tumor, and circadian clock protein PERIOD2 (PER2) acts as a tumor suppressor in cancer; however, little is known about its involvement in chemosensitivity. Methods This study aimed to investigate the role and underlying mechanisms of PER2 in ovarian cancer sensitivity to cisplatin. Overexpression and knockdown of PER2 were performed to explore its role in ovarian cancer cell sensitivity to cisplatin both in vitro and in vivo. The protein levels of PI3K, AKT, caspase 3, E-cadherin, and other drug resistance-related molecules were determined in parental SKOV3 and SKOV3/DDP cells as well as in xenograft tumor tissues. Results Compared with parental cells, SKOV3/DDP cells had dramatically decreased PER2 expression, possibly due to hypermethylation in the PER2 promoter. PER2 overexpression significantly inhibited proliferation while promoting cisplatin-induced apoptosis in SKOV3 and SKOV3/DDP cells. In agreement, PER2-overexpressing SKOV3/DPP cells yielded significantly reduced tumor mass in cisplatin-treated mice compared with control cells. Mechanistically, PER2 overexpression remarkably reduced the protein amounts of PI3K, AKT, and MDR1 while increasing those of caspase 3 and E-cadherin in tumor tissues. Knockdown of PER2 exhibited opposite effects. PER2 overexpression also reduced the serum levels of TNF-α and IL-6 in tumor-bearing mice before the initiation of cisplatin treatment. Conclusion This study suggests that loss of PER2 contributes to cisplatin resistance in SKOV3 cells, possibly by activating the PI3K/AKT pathway and EMT, inhibiting apoptosis, and promoting drug efflux and inflammatory responses. Overexpression of PER2 could reverse these alterations and sensitize both parental SKOV3 and SKOV3/DDP cells to cisplatin.
Collapse
Affiliation(s)
- Zhaoxia Wang
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Fengyan Li
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Meiyan Wei
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Sanyuan Zhang
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Tong Wang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| |
Collapse
|
46
|
Mechanisms of tRNA-derived fragments and tRNA halves in cancer treatment resistance. Biomark Res 2020; 8:52. [PMID: 33072328 PMCID: PMC7559774 DOI: 10.1186/s40364-020-00233-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/02/2020] [Indexed: 12/22/2022] Open
Abstract
The tRNA-derived fragments (tRFs) and tRNA halves (tiRNAs) are newly discovered noncoding RNAs in recent years. They are derived from specific cleavage of mature and pre-tRNAs and expressed in various cancers. They enhance cell proliferation and metastasis or inhibit cancer progression. Many studies have investigated their roles in the diagnosis, progression, metastasis, and prognosis of various cancers, but the mechanisms through which they are involved in resistance to cancer treatment are unclear. This review outlines the classification of tRFs and tiRNAs and their mechanisms in cancer drug resistance, thus providing new ideas for cancer treatment.
Collapse
|
47
|
Dai L, Song K, Di W. Adipocytes: active facilitators in epithelial ovarian cancer progression? J Ovarian Res 2020; 13:115. [PMID: 32967712 PMCID: PMC7513299 DOI: 10.1186/s13048-020-00718-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022] Open
Abstract
There is growing evidence that adipocytes play important roles in the progression of multiple cancers. Moreover, in obesity, adipocytes alter their original functions and contribute to the metabolic and inflammatory changes of adipose tissue microenvironment, which can further enhance tumor development. At present, the roles of adipocytes in the pathogenesis of epithelial ovarian cancer (EOC) are far from being fully elucidated. Herein, we summarized the recent advances in understanding the roles of adipocytes in EOC progression. Adipocytes, close neighbors of EOC tissue, promote EOC growth, invasion, metastasis and angiogenesis through adipokine secretion, metabolic remodeling and immune microenvironment modulation. Moreover, adipocytes are important therapeutic targets and may work as useful anticancer drug delivery depot for EOC treatment. Furthermore, adipocytes also act as a therapeutic obstacle for their involvement in EOC treatment resistance. Hence, better characterization of the adipocytes in EOC microenvironment and the crosstalk between adipocytes and EOC cells may provide insights into EOC progression and suggest novel therapeutic opportunities.
Collapse
Affiliation(s)
- Lan Dai
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China. .,Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Keqi Song
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wen Di
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China. .,Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China. .,State Key Laboratory of Oncogene and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
48
|
Beyranvand Nejad E, Labrie C, van der Sluis TC, van Duikeren S, Franken KLMC, Roosenhoff R, Arens R, van Hall T, van der Burg SH. Interleukin-6-mediated resistance to immunotherapy is linked to impaired myeloid cell function. Int J Cancer 2020; 148:211-225. [PMID: 32875568 PMCID: PMC7693233 DOI: 10.1002/ijc.33280] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/30/2020] [Accepted: 08/14/2020] [Indexed: 12/17/2022]
Abstract
High serum levels of interleukin-6 (IL-6) correlate with poor prognosis and chemotherapy resistance in several cancers. The underlying mechanisms and its effects on immunotherapy are largely unknown. To address this, we developed a human papillomavirus type 16 (HPV16)-associated tumor model expressing IL-6 to investigate the impact of tumor-expressed IL-6 during cisplatin chemotherapy and HPV16 synthetic long peptide vaccination as immunotherapy. The effects of tumor-produced IL-6 on tumor growth, survival and the tumor microenvironment were analyzed. Our data demonstrated that tumor-produced IL-6 conferred resistance to cisplatin and therapeutic vaccination. This was not caused by a changed in vitro or in vivo growth rate of tumor cells, or a changed sensitivity of tumor cells to chemotherapy or T-cell-mediated killing. Furthermore, no overt differences in the frequencies of tumor-infiltrating subsets of T cells or CD11b+ myeloid cells were observed. IL-6, however, affected the systemic and local function of myeloid cells, reflected by a strong reduction of major histocompatibility complex (MHC) class II expression on all major myeloid cell subtypes. Resistance to both therapies was associated with a changed intratumoral influx of MHC class II+ myeloid cells toward myeloid cells with no or lower MHC class II expression. Importantly, while these IL-6-mediated effects provided resistance to the immunotherapy and chemotherapy as single therapies, their combination still successfully mediated tumor control. In conclusion, IL-6-mediated therapy resistance is caused by an extrinsic mechanism involving an impaired function of intratumoral myeloid cells. The fact that resistance can be overcome by combination therapies provides direction to more effective therapies for cancer.
Collapse
Affiliation(s)
- Elham Beyranvand Nejad
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Camilla Labrie
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Tetje C van der Sluis
- Department of Immunology, Leiden University Medical Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne van Duikeren
- Department of Immunology, Leiden University Medical Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Kees L M C Franken
- Department of Immunology, Leiden University Medical Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Rueshandra Roosenhoff
- Department of Immunology, Leiden University Medical Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Ramon Arens
- Department of Immunology, Leiden University Medical Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
49
|
Barillé-Nion S, Lohard S, Juin PP. Targeting of BCL-2 Family Members during Anticancer Treatment: A Necessary Compromise between Individual Cell and Ecosystemic Responses? Biomolecules 2020; 10:E1109. [PMID: 32722518 PMCID: PMC7464802 DOI: 10.3390/biom10081109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 01/07/2023] Open
Abstract
The imbalance between BCL-2 homologues and pro-death counterparts frequently noted in cancer cells endows them with a cell autonomous survival advantage. To eradicate ectopic cells, inhibitors of these homologues (BH3 mimetics) were developed to trigger, during anticancer treatment, full activation of the canonical mitochondrial apoptotic pathway and related caspases. Despite efficiency in some clinical settings, these compounds do not completely fulfill their initial promise. We herein put forth that a growing body of evidence indicates that mitochondrial integrity, controlled by BCL-2 family proteins, and downstream caspases regulate other cell death modes and influence extracellular signaling by committed cells. Moreover, intercellular communications play a key role in spreading therapeutic response across cancer cell populations and in engaging an immune response. We thus advocate that BH3 mimetics administration would be more efficient in the long term if it did not induce apoptosis in all sensitive cells at the same time, but if it could instead allow (or trigger) death signal production by non-terminally committed dying cell populations. The development of such a trade-off strategy requires to unravel the effects of BH3 mimetics not only on each individual cancer cell but also on homotypic and heterotypic cell interactions in dynamic tumor ecosystems.
Collapse
Affiliation(s)
- Sophie Barillé-Nion
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERMU1232, Université de Nantes, F-44000 Nantes, France; (S.B.-N.); (S.L.)
- SIRIC ILIAD, 44000 Nantes, France
| | - Steven Lohard
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERMU1232, Université de Nantes, F-44000 Nantes, France; (S.B.-N.); (S.L.)
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Philippe P. Juin
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERMU1232, Université de Nantes, F-44000 Nantes, France; (S.B.-N.); (S.L.)
- SIRIC ILIAD, 44000 Nantes, France
- Institut de Cancérologie de l’Ouest, 15 Rue André Boquel, 49055 Angers, France
| |
Collapse
|
50
|
Zhu Y, Zhang G, Li M, Ma L, Huang J, Qiu L. Ultrasound-Augmented Phase Transition Nanobubbles for Targeted Treatment of Paclitaxel-Resistant Cancer. Bioconjug Chem 2020; 31:2008-2020. [PMID: 32628454 DOI: 10.1021/acs.bioconjchem.0c00364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yi Zhu
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Ultrasound, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Guonan Zhang
- Department of Gynecological Oncology, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Meiying Li
- Department of Biochemistry & Molecular Biology, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Lang Ma
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianming Huang
- Department of Biochemistry & Molecular Biology, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Li Qiu
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|