51
|
Lin X, Yoshikawa N, Liu W, Matsukawa T, Nakamura K, Yoshihara M, Koya Y, Sugiyama M, Tamauchi S, Ikeda Y, Yokoi A, Shimizu Y, Kajiyama H. DDIT4 Facilitates Lymph Node Metastasis via the Activation of NF-κB Pathway and Epithelial-Mesenchymal Transition. Reprod Sci 2023; 30:2829-2841. [PMID: 37016173 DOI: 10.1007/s43032-023-01230-y] [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: 10/25/2022] [Accepted: 03/27/2023] [Indexed: 04/06/2023]
Abstract
This study was aimed to identify a novel metastasis-promoting molecule and elucidate its functional and prognostic roles in cervical cancer. DDIT4 (DNA-damage-inducible transcript 4), a hypoxia-inducible gene, was identified by analyzing multiple microarray databases. The correlation between DDIT4 expression in immunohistochemistry and clinicopathological characteristics in the public database and our cohort was evaluated by statistical analysis. Transwell® assay and wound-healing assay to determine cell migration and invasion were performed. DDIT4 was knocked down using siRNA or lentiviral vectors. The potential downstream pathways of DDIT4 were explored and verified by a gene set enrichment analysis and western blotting. The in vivo metastatic capability was determined with the use of an intraperitoneal injection mouse model. In the analysis of the public database and our cohort, DDIT4 high expression was significantly related to short overall survival and lymph node metastasis in patients with early-stage cervical cancer. The knockdown of DDIT4 attenuated the migration and invasion activity of tumor cells in vitro and reduced the expression of epithelial-mesenchymal transition (EMT)-related proteins and the NF-κB pathway in cervical cancer cells. DDIT4 also promoted tumor progression in the mouse model. Our results indicate that DDIT4 can be a prognostic indicator in cervical cancer and promote lymph node metastasis, augmenting malignancy via the EMT and NF-kB pathways.
Collapse
Affiliation(s)
- Xinxin Lin
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| | - Wenting Liu
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Matsukawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Kae Nakamura
- Center for Low-Temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Yoshihiro Koya
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mai Sugiyama
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Tamauchi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Yoshiki Ikeda
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Yusuke Shimizu
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| |
Collapse
|
52
|
Jena SK, Das S, Chakraborty S, Ain R. Molecular determinants of epithelial mesenchymal transition in mouse placenta and trophoblast stem cell. Sci Rep 2023; 13:10978. [PMID: 37414855 PMCID: PMC10325982 DOI: 10.1038/s41598-023-37977-2] [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: 03/22/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023] Open
Abstract
Trophectoderm cells of the blastocyst are the precursor of the placenta that is comprised of trophoblast, endothelial and smooth muscle cells. Since trophoectoderm cells are epithelial in nature, epithelial mesenchymal transition (EMT) of trophoblast stem (TS) cells might play pivotal role in placental morphogenesis. However, the molecular regulation of EMT during placental development and trophoblast differentiation still remained elusive. In this report, we sought to identify the molecular signature that regulates EMT during placental development and TS cell differentiation in mice. On E7.5 onwards the TS cells, located in the ectoplacental cone (EPC), rapidly divide and differentiate leading to formation of placenta proper. Using a real time PCR based array of functional EMT transcriptome with RNA from mouse implantation sites (IS) on E7.5 and E9.5, it was observed that there was an overall reduction of EMT gene expression in the IS as gestation progressed from E7.5 to E9.5 albeit the levels of EMT gene expression were substantial on both days. Further validation of array results using real time PCR and western blot analysis showed significant decrease in EMT-associated genes that included (a) transcription factors (Snai2, Zeb1, Stat3 and Foxc2), (b) extracellular matrix and cell adhesion related genes (Bmp1, Itga5, Vcan and Col3A1), (c) migration and motility- associated genes (Vim, Msn and FN1) and (d) differentiation and development related genes (Wnt5b, Jag1 and Cleaved Notch-1) on E9.5. To understand whether EMT is an ongoing process during placentation, the EMT-associated signatures genes, prevalent on E 7.5 and 9.5, were analysed on E12.5, E14.5 and E17.5 of mouse placenta. Interestingly, expression of these EMT-signature proteins were significantly higher at E12.5 though substantial expressions was observed in placenta with progression of gestation from mid- to late. To evaluate whether TS cells have the potential to undergo EMT ex vivo, TS cells were subjected to EMT induction, which was confirmed using morphological analysis and marker gene expression. Induction of EMT in TS cells showed similar gene expression profile of placental EMT. These results have broad biological implications, as inadequate mesenchymal transition leading to improper trophoblast-vasculogenic mimicry leads to placental pathophysiology and pregnancy failure.
Collapse
Affiliation(s)
- Shipra Kanti Jena
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta, West Bengal, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, UP, 201002, India
| | - Shreya Das
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta, West Bengal, 700032, India
| | - Shreeta Chakraborty
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta, West Bengal, 700032, India
| | - Rupasri Ain
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta, West Bengal, 700032, India.
- Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, UP, 201002, India.
| |
Collapse
|
53
|
Lei S, Du X, Tan K, He X, Zhu Y, Zhao S, Yang Z, Dou G. CRP‑1 promotes the malignant behavior of hepatocellular carcinoma cells via activating epithelial‑mesenchymal transition and Wnt/β‑catenin signaling. Exp Ther Med 2023; 26:314. [PMID: 37273753 PMCID: PMC10236095 DOI: 10.3892/etm.2023.12013] [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: 12/28/2022] [Accepted: 04/18/2023] [Indexed: 06/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. It has been reported that cysteine rich protein 1 (CRP-1) is dysregulated in several types of human cancer; however, its role in HCC is poorly understood. Therefore, the current study aimed to investigate the role of CRP-1 in HCC. Western blotting and reverse transcription-quantitative PCR results showed that CRP-1 was upregulated in HCC cell lines. Furthermore, for in vitro experiments, CRP-1 was knocked down and overexpressed in the HCC cell lines Hep 3B2.1-7 and BEL-7405, respectively. c-Myc and proliferating cell nuclear antigen upregulation, and cleaved caspase 3 and poly(ADP-ribose) polymerase downregulation suggested that CRP-1 silencing could inhibit the proliferation and colony-forming ability of HCC cells, and induce apoptosis. In addition, CRP-1 overexpression promoted the malignant behavior of HCC cells and induced epithelial-mesenchymal transition (EMT), as verified by E-cadherin downregulation, and N-cadherin and vimentin upregulation. Additionally, CRP-1 overexpression promoted the nuclear translocation of β-catenin, and activated the expression of cyclin D1 and matrix metalloproteinase-7. Furthermore, inhibition of Wnt/β-catenin signaling, following cell treatment with XAV-939, an inhibitor of the Wnt/β-catenin signaling pathway, abrogated the effects of CRP-1 on enhancing the proliferation and migration of HCC cells. These findings indicated that the regulatory effect of CRP-1 on HCC cells could be mediated by the Wnt/β-catenin signaling pathway. Overall, CRP-1 could promote the proliferation and migration of HCC cell lines, partially via promoting EMT and activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Shixiong Lei
- Department of Interventional Medicine, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Xilin Du
- Department of General Surgery, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Kai Tan
- Department of General Surgery, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Xiaojun He
- Department of General Surgery, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yejing Zhu
- Department of General Surgery, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Shoujie Zhao
- Department of General Surgery, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Zhenyu Yang
- Department of General Surgery, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Gang Dou
- Department of General Surgery, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
54
|
Si H, Esquivel M, Mendoza Mendoza E, Roarty K. The covert symphony: cellular and molecular accomplices in breast cancer metastasis. Front Cell Dev Biol 2023; 11:1221784. [PMID: 37440925 PMCID: PMC10333702 DOI: 10.3389/fcell.2023.1221784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer has emerged as the most commonly diagnosed cancer and primary cause of cancer-related deaths among women worldwide. Although significant progress has been made in targeting the primary tumor, the effectiveness of systemic treatments to prevent metastasis remains limited. Metastatic disease continues to be the predominant factor leading to fatality in the majority of breast cancer patients. The existence of a prolonged latency period between initial treatment and eventual recurrence in certain patients indicates that tumors can both adapt to and interact with the systemic environment of the host, facilitating and sustaining the progression of the disease. In order to identify potential therapeutic interventions for metastasis, it will be crucial to gain a comprehensive framework surrounding the mechanisms driving the growth, survival, and spread of tumor cells, as well as their interaction with supporting cells of the microenvironment. This review aims to consolidate recent discoveries concerning critical aspects of breast cancer metastasis, encompassing the intricate network of cells, molecules, and physical factors that contribute to metastasis, as well as the molecular mechanisms governing cancer dormancy.
Collapse
Affiliation(s)
- Hongjiang Si
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Madelyn Esquivel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Erika Mendoza Mendoza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Kevin Roarty
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| |
Collapse
|
55
|
Javed AA, Floortje van Oosten A, Habib JR, Hasanain A, Kinny-Köster B, Gemenetzis G, Groot VP, Ding D, Cameron JL, Lafaro KJ, Burns WR, Burkhart RA, Yu J, He J, Wolfgang CL. A Delay in Adjuvant Therapy Is Associated With Worse Prognosis Only in Patients With Transitional Circulating Tumor Cells After Resection of Pancreatic Ductal Adenocarcinoma. Ann Surg 2023; 277:866-872. [PMID: 36111839 DOI: 10.1097/sla.0000000000005710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The aim of the study was to assess the association of circulating tumor cells (CTCs) with survival as a biomarker in pancreatic ductal adenocarcinoma (PDAC) within the context of a delay in the initiation of adjuvant therapy. BACKGROUND Outcomes in patients with PDAC remain poor and are driven by aggressive systemic disease. Although systemic therapies improve survival in resected patients, factors such as a delay in the initiation of adjuvant therapy are associated with worse outcomes. CTCs have previously been shown to be predictive of survival. METHODS A retrospective study was performed on PDAC patients enrolled in the prospective CircuLating tUmor cellS in pancreaTic cancER trial (NCT02974764) on CTC-dynamics at the Johns Hopkins Hospital. CTCs were isolated based on size (isolation by size of epithelial tumor cells; Rarecells) and counted and characterized by subtype using immunofluorescence. The preoperative and postoperative blood samples were used to identify 2 CTC types: epithelial CTCs (eCTCs), expressing pancytokeratin, and transitional CTCs (trCTCs), expressing both pancytokeratin and vimentin. Patients who received adjuvant therapy were compared with those who did not. A delay in the receipt of adjuvant therapy was defined as the initiation of therapy ≥8 weeks after surgical resection. Clinicopathologic features, CTCs characteristics, and outcomes were analyzed. RESULTS Of 101 patients included in the study, 43 (42.5%) experienced a delay in initiation and 20 (19.8%) did not receive adjuvant therapy. On multivariable analysis, the presence of trCTCs ( P =0.002) and the absence of adjuvant therapy ( P =0.032) were associated with worse recurrence-free survival (RFS). Postoperative trCTC were associated with poorer RFS, both in patients with a delay in initiation (12.4 vs 17.9 mo, P =0.004) or no administration of adjuvant chemotherapy (3.4 vs NR, P =0.016). However, it was not associated with RFS in patients with timely initiation of adjuvant chemotherapy ( P =0.293). CONCLUSIONS Postoperative trCTCs positivity is associated with poorer RFS only in patients who either experience a delay in initiation or no receipt of adjuvant therapy. This study suggests that a delay in the initiation of adjuvant therapy could potentially provide residual systemic disease (trCTCs) a window of opportunity to recover from the surgical insult. Future studies are required to validate these findings and explore the underlying mechanisms involved.
Collapse
Affiliation(s)
- Ammar A Javed
- Department of Surgery, New York University Langone Hospital, New York City, NY
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Anne Floortje van Oosten
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Utrecht University, The Netherlands
| | - Joseph R Habib
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alina Hasanain
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Benedict Kinny-Köster
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Georgios Gemenetzis
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Vincent P Groot
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ding Ding
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/ Northwell, Manhasset, NY
| | - John L Cameron
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kelly J Lafaro
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - William R Burns
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Richard A Burkhart
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jun Yu
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jin He
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | | |
Collapse
|
56
|
Gopikrishnan M, R HC, R G, Ashour HM, Pintus G, Hammad M, Kashyap MK, C GPD, Zayed H. Therapeutic and diagnostic applications of exosomal circRNAs in breast cancer. Funct Integr Genomics 2023; 23:184. [PMID: 37243750 DOI: 10.1007/s10142-023-01083-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 05/29/2023]
Abstract
Circular RNAs (circRNAs) are regulatory elements that are involved in orchestrating gene expression and protein functions and are implicated in various biological processes including cancer. Notably, breast cancer has a significant mortality rate and is one of the most common malignancies in women. CircRNAs have been demonstrated to contribute to the pathogenesis of breast cancer including its initiation, progression, metastasis, and resistance to drugs. By acting as miRNA sponges, circRNAs can indirectly influence gene expression by disrupting miRNA regulation of their target genes, ultimately altering the course of cancer development and progression. Additionally, circRNAs can interact with proteins and modulate their functions including signaling pathways involved in the initiation and development of cancer. Recently, circRNAs can encode peptides that play a role in the pathophysiology of breast cancer and other diseases and their potential as diagnostic biomarkers and therapeutic targets for various cancers including breast cancer. CircRNAs possess biomarkers that differentiate, such as stability, specificity, and sensitivity, and can be detected in several biological specimens such as blood, saliva, and urine. Moreover, circRNAs play an important role in various cellular processes including cell proliferation, differentiation, and apoptosis, all of which are integral factors in the development and progression of cancer. This review synthesizes the functions of circRNAs in breast cancer, scrutinizing their contributions to the onset and evolution of the disease through their interactions with exosomes and cancer-related intracellular pathways. It also delves into the potential use of circRNA as a biomarker and therapeutic target against breast cancer. It discusses various databases and online tools that offer crucial circRNA information and regulatory networks. Lastly, the challenges and prospects of utilizing circRNAs in clinical settings associated with breast cancer are explored.
Collapse
Affiliation(s)
- Mohanraj Gopikrishnan
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| | - Hephzibah Cathryn R
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| | - Gnanasambandan R
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| | - Hossam M Ashour
- Department of Integrative Biology, College of Arts and Sciences, University of South Florida, St. Petersburg, Florida, 33701, USA
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Mohamed Hammad
- Department of Stem Cell Biology and Regenerative Medicine, City of Hope Beckman Research Institute, Duarte, California, USA
| | - Manoj Kumar Kashyap
- Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Manesar (Gurugram), Panchgaon, Haryana (HR), 122413, India
- Clinical Biosamples & Research Services (CBRS), Noida, Uttar Pradesh, 201301, India
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India.
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, 2713, Doha, Qatar.
| |
Collapse
|
57
|
Cai X, Li S, Zeng X, Xu M, Wang Z, Singhi AD, Tang D, Li S, Yates NA, Yang D, Xie W. Inhibition of the SLC35B2-TPST2 Axis of Tyrosine Sulfation Attenuates the Growth and Metastasis of Pancreatic Ductal Adenocarcinom. Cell Mol Gastroenterol Hepatol 2023; 16:473-495. [PMID: 37192689 PMCID: PMC10393550 DOI: 10.1016/j.jcmgh.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer deaths in the United States. Tyrosine sulfation, catalyzed by the tyrosylprotein sulfotransferase 2 (TPST2), is a post-translational modification essential for protein-protein interactions and cellular functions. Solute carrier family 35 member B (SLC35B2) is a key transporter that transports the universal sulfate donor 3'-phosphoadenosine 5'-phosphosulfate into the Golgi apparatus where the protein sulfation occurs. The goal of this study was to determine whether and how the SLC35B2-TPST2 axis of tyrosine sulfation plays a role in PDAC. METHODS Gene expression was analyzed in PDAC patients and mice. Human PDAC MIA PaCa-2 and PANC-1 cells were used for in vitro studies. TPST2-deficient MIA PaCa-2 cells were generated to assess xenograft tumor growth in vivo. Mouse PDAC cells derived from the KrasLSL-G12D/+;Tp53L/+;Pdx1-Cre (KPC) mice were used to generate Tpst2 knockout KPC cells to evaluate tumor growth and metastasis in vivo. RESULTS High expressions of SLC35B2 and TPST2 were correlated with poor PDAC patient survival. Knocking down SLC35B2 or TPST2, or pharmacologicically inhibiting sulfation, resulted in the inhibition of PDAC cell proliferation and migration in vitro. TPST2-deficient MIA PaCa-2 cells showed inhibited xenograft tumor growth. Orthotopic inoculation of Tpst2 knockout KPC cells in mice showed inhibition of primary tumor growth, local invasion, and metastasis. Mechanistically, the integrin β4 was found to be a novel substrate of TPST2. Inhibition of sulfation destabilizes integrin β4 protein, which may have accounted for the suppression of metastasis. CONCLUSIONS Targeting the SLC35B2-TPST2 axis of tyrosine sulfation may represent a novel approach for therapeutic intervention of PDAC.
Collapse
Affiliation(s)
- Xinran Cai
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sihan Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xuemei Zeng
- Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, Pennsylvania
| | - Meishu Xu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zehua Wang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Aatur D Singhi
- Department of Pathology, School of Medicine, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, School of Medicine, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daolin Tang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nathan A Yates
- Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, Pennsylvania; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Da Yang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
58
|
Serrano-Rivero Y, Salazar-Uribe J, Rubio-Carrasquilla M, Camacho-Casanova F, Sánchez-Ramos O, González-Pose A, Moreno E. Selecting Nanobodies Specific for the Epidermal Growth Factor from a Synthetic Nanobody Library. Molecules 2023; 28:molecules28104043. [PMID: 37241784 DOI: 10.3390/molecules28104043] [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: 03/24/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The epidermal growth factor (EGF) is one of the most critical ligands of the EGF receptor (EGFR), a well-known oncogene frequently overexpressed in cancerous cells and an important therapeutic target in cancer. The EGF is the target of a therapeutic vaccine aimed at inducing an anti-EGF antibody response to sequester this molecule from serum. However, strikingly, very few investigations have focused on EGF immunotargeting. Since the use of nanobodies (Nbs) for EGF neutralization may be an effective therapeutic strategy in several types of cancer, in this study, we decided to generate anti-EGF Nbs from a recently constructed, phage-displaying synthetic nanobody library. To our knowledge, this is the first attempt to obtain anti-EGF Nbs from a synthetic library. By applying a selection strategy that uses four different sequential elution steps along with three rounds of selection, we obtained four different EGF-specific Nb clones, and also tested their binding capabilities as recombinant proteins. The obtained results are very encouraging and demonstrate the feasibility of selecting nanobodies against small antigens, such as the EGF, from synthetic libraries.
Collapse
Affiliation(s)
| | | | | | - Frank Camacho-Casanova
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | - Oliberto Sánchez-Ramos
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | - Alaín González-Pose
- Faculty of Basic Sciences, University of Medellin, Medellin 050026, Colombia
| | - Ernesto Moreno
- Faculty of Basic Sciences, University of Medellin, Medellin 050026, Colombia
| |
Collapse
|
59
|
Jun X, Gao S, Yu L, Wang G. The clinical relevance and prediction efficacy from therapy of tumor microenvironment related signature score in colorectal cancer. Front Oncol 2023; 13:1123455. [PMID: 37234984 PMCID: PMC10207322 DOI: 10.3389/fonc.2023.1123455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/18/2023] [Indexed: 05/28/2023] Open
Abstract
Introduction As the top 3 cancer in terms of incidence and mortality, the first-line treatment for CRC includes FOLFOX, FOLFIRI, Cetuximab or immunotherapy. However, the drug sensitivity of patients to regimens is different. There has been increasing evidence that immune components of TME can affect the sensitivity of patients to drugs. Therefore, it is necessary to define novo molecular subtypes of CRC based on TME immune components, and screen patients who are sensitive to the treatments, to make personalized therapy possible. Methods We analyzed the expression profiles and 197 TME-related signatures of 1775 patients using ssGSEA, univariate Cox proportional risk model and LASSO-Cox regression model, and defined a novo molecular subtype (TMERSS) of CRC. Simultaneously, we compared the clinicopathological factors, antitumor immune activity, immune cell abundance and differences of cell states in different TMERSS subtypes. In addition, patients sensitive to the therapy were screened out by correlation analysis between TMERSS subtypes and drug responses. Results Compared with low TMERSS subtype, high TMERSS subtype has a better outcome, which may be associated to higher abundance of antitumor immune cell in high TMERSS subtype. Our findings suggested that the high TMERSS subtype may have a higher proportion of respondents to Cetuximab agent and immunotherapy, while the low TMERSS subtype may be more suitable for treatment with FOLFOX and FOLFIRI regimens. Discussion In conclusion, the TMERSS model may provide a partial reference for the prognosis evaluation of patients, the prediction of drug sensitivity, and the implementation of clinical decision-making.
Collapse
Affiliation(s)
- Xiang Jun
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shengnan Gao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Yu
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guiyu Wang
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
60
|
Asghari S, Mahmoudifard M. The detection of the captured circulating tumor cells on the core-shell nanofibrous membrane using hyaluronic acid-functionalized graphene quantum dots. J Biomed Mater Res B Appl Biomater 2023; 111:1121-1132. [PMID: 36727427 DOI: 10.1002/jbm.b.35219] [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: 05/18/2022] [Revised: 11/26/2022] [Accepted: 12/23/2022] [Indexed: 02/03/2023]
Abstract
In recent years, cancerous cases have increased remarkably worldwide, and metastasis is the leading cause of death. Therefore, research on the early detection of cancer and metastasis has expanded to aid successful cancer treatment. Here in this paper, at the first step, an electrospun nanofibrous membrane (NFM) with a core-shell structure was fabricated from PCL and HA to achieve cancer cell capturing (about 75% of cells). On the other hand, hyaluronic acid (HA)-functionalized graphene quantum dots (GQDs) were used to detect captured cancer cells on NFM through the changes in photoluminescence intensity. Therefore, CD44 receptor-HA interaction is the main principle used for both entrapment and detection of cancer cells. Results demonstrated the GQD-HA fluorescent intensity of solution decreased through the increase of the captured cancer cell numbers on NFM, which is related to the more adsorption of GQD nanocomposites to the CD44 receptors. In contrast, this intensity for noncancerous cells was steady with any cell concentrations. This difference shows the system's remarkable selectivity and specificity, which can be crucial in fluorescent imaging for accurate cancer diagnosis.
Collapse
Affiliation(s)
- Sahar Asghari
- Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Matin Mahmoudifard
- Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
61
|
Lopez-Cavestany M, Hahn SB, Hope JM, Reckhorn NT, Greenlee JD, Schwager SC, VanderBurgh JA, Reinhart-King CA, King MR. Matrix stiffness induces epithelial-to-mesenchymal transition via Piezo1-regulated calcium flux in prostate cancer cells. iScience 2023; 26:106275. [PMID: 36950111 PMCID: PMC10025097 DOI: 10.1016/j.isci.2023.106275] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 01/21/2023] [Accepted: 02/18/2023] [Indexed: 02/27/2023] Open
Abstract
Cells utilize calcium channels as one of the main signaling mechanisms to sense changes in the extracellular space and convert these changes to intracellular signals. Calcium regulates several key signaling networks, such as the induction of EMT. The current study expands on the understanding of how EMT is controlled via the mechanosensitive calcium channel Piezo1 in cancerous cells, which senses changes in the extracellular matrix stiffness. We model the biophysical environment of healthy and cancerous prostate tissue using polyacrylamide gels of different stiffnesses. Significant increases in calcium steady-state concentration, vimentin expression, and aspect ratio, and decreases in E-cadherin expression were observed by increasing matrix stiffness and also after treatment with Yoda1, a chemical agonist of Piezo1. Overall, this study concludes that Piezo1-regulated calcium flux plays a role in prostate cancer cell metastatic potential by sensing changes in ECM stiffness and modulating EMT markers.
Collapse
Affiliation(s)
- Maria Lopez-Cavestany
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Su Bin Hahn
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Jacob M. Hope
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Noah T. Reckhorn
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Joshua D. Greenlee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Samantha C. Schwager
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Jacob A. VanderBurgh
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | | | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
62
|
Kahm YJ, Kim IG, Kim RK. RanBP1: A Potential Therapeutic Target for Cancer Stem Cells in Lung Cancer and Glioma. Int J Mol Sci 2023; 24:ijms24076855. [PMID: 37047826 PMCID: PMC10095367 DOI: 10.3390/ijms24076855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
Cancer stem cells (CSCs) are known to be one of the factors that make cancer treatment difficult. Many researchers are thus conducting research to efficiently destroy CSCs. Therefore, we sought to suggest a new target that can efficiently suppress CSCs. In this study, we observed a high expression of Ran-binding protein 1 (RanBP1) in lung cancer stem cells (LCSCs) and glioma stem cells (GSCs). Upregulated RanBP1 expression is strongly associated with the expression of CSC marker proteins and CSC regulators. In addition, an elevated RanBP1 expression is strongly associated with a poor patient prognosis. CSCs have the ability to resist radiation, and RanBP1 regulates this ability. RanBP1 also affects the metastasis-associated epithelial–mesenchymal transition (EMT) phenomenon. EMT marker proteins and regulatory proteins are affected by RanBP1 expression, and cell motility was regulated according to RanBP1 expression. The cancer microenvironment influences cancer growth, metastasis, and cancer treatment. RanBP1 can modulate the cancer microenvironment by regulating the cytokine IL-18. Secreted IL-18 acts on cancer cells and promotes cancer malignancy. Our results reveal, for the first time, that RanBP1 is an important regulator in LCSCs and GSCs, suggesting that it holds potential for use as a potential therapeutic target.
Collapse
Affiliation(s)
- Yeon-Jee Kahm
- Department of Radiation Biology, Environmental Safety Assessment Research Division, Korea Atomic Energy Research Institute, Yuseong-gu, Daejeon 34057, Republic of Korea
- Department of Radiation Science and Technology, Korea University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - In-Gyu Kim
- Department of Radiation Biology, Environmental Safety Assessment Research Division, Korea Atomic Energy Research Institute, Yuseong-gu, Daejeon 34057, Republic of Korea
- Department of Radiation Science and Technology, Korea University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Rae-Kwon Kim
- Department of Radiation Biology, Environmental Safety Assessment Research Division, Korea Atomic Energy Research Institute, Yuseong-gu, Daejeon 34057, Republic of Korea
- Department of Radiation Science and Technology, Korea University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| |
Collapse
|
63
|
Fang T, Jiao Z, You Y, Cao J, Wang C, Liu J, Zhao W. Lenvatinib inhibited HCC cell migration and invasion through regulating the transcription and ubiquitination of UHRF1 and DNMT1. Biochem Pharmacol 2023; 210:115489. [PMID: 36893815 DOI: 10.1016/j.bcp.2023.115489] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common causes of malignancy-related deaths. Lenvatinib, as a multi-targeted tyrosine kinase inhibitor, has gained increasing attention for its antitumor activity. However, the effect and mechanisms of Lenvatinib on HCC metastasis are virtually unknown. In this study, we revealed that Lenvatinib inhibited HCC cell motility and epithelial mesenchymal transition (EMT), along with cell adhesion and extension. Concomitant high DNMT1 and UHRF1 mRNA levels were in HCC patients and indicated worse prognosis. On the one hand, Lenvatinib modulated the transcription of UHRF1 and DNMT1via negatively regulation of ERK/MAPK pathway. On the other hand, Lenvatinib downregulated DNMT1 and UHRF1 expression by promoting their protein degradation through ubiquitin-proteasome pathway, consequently, resulting in upregulation of E-Cadherin. Moreover, Lenvatinib attenuated Huh7 cell adhesion and metastasis in vivo. Our findings provided insight into the intriguing molecular mechanisms regarding the anti-metastasis effect of Lenvatinib in HCC.
Collapse
Affiliation(s)
- Ting Fang
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, PR China
| | - Zhen Jiao
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, PR China
| | - Yuting You
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, PR China
| | - Jiahao Cao
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, PR China
| | - Chuanzheng Wang
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, PR China
| | - Jingjing Liu
- Xiamen Key Laboratory of Intestinal Microbiome and Human Health, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, PR China.
| | - Wenxiu Zhao
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, PR China.
| |
Collapse
|
64
|
Wu Y, Terekhanova NV, Caravan W, Naser Al Deen N, Lal P, Chen S, Mo CK, Cao S, Li Y, Karpova A, Liu R, Zhao Y, Shinkle A, Strunilin I, Weimholt C, Sato K, Yao L, Serasanambati M, Yang X, Wyczalkowski M, Zhu H, Zhou DC, Jayasinghe RG, Mendez D, Wendl MC, Clark D, Newton C, Ruan Y, Reimers MA, Pachynski RK, Kinsinger C, Jewell S, Chan DW, Zhang H, Chaudhuri AA, Chheda MG, Humphreys BD, Mesri M, Rodriguez H, Hsieh JJ, Ding L, Chen F. Epigenetic and transcriptomic characterization reveals progression markers and essential pathways in clear cell renal cell carcinoma. Nat Commun 2023; 14:1681. [PMID: 36973268 PMCID: PMC10042888 DOI: 10.1038/s41467-023-37211-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
Identifying tumor-cell-specific markers and elucidating their epigenetic regulation and spatial heterogeneity provides mechanistic insights into cancer etiology. Here, we perform snRNA-seq and snATAC-seq in 34 and 28 human clear cell renal cell carcinoma (ccRCC) specimens, respectively, with matched bulk proteogenomics data. By identifying 20 tumor-specific markers through a multi-omics tiered approach, we reveal an association between higher ceruloplasmin (CP) expression and reduced survival. CP knockdown, combined with spatial transcriptomics, suggests a role for CP in regulating hyalinized stroma and tumor-stroma interactions in ccRCC. Intratumoral heterogeneity analysis portrays tumor cell-intrinsic inflammation and epithelial-mesenchymal transition (EMT) as two distinguishing features of tumor subpopulations. Finally, BAP1 mutations are associated with widespread reduction of chromatin accessibility, while PBRM1 mutations generally increase accessibility, with the former affecting five times more accessible peaks than the latter. These integrated analyses reveal the cellular architecture of ccRCC, providing insights into key markers and pathways in ccRCC tumorigenesis.
Collapse
Affiliation(s)
- Yige Wu
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Nadezhda V Terekhanova
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Wagma Caravan
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Nataly Naser Al Deen
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Preet Lal
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Siqi Chen
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Chia-Kuei Mo
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Song Cao
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Yize Li
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Alla Karpova
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Ruiyang Liu
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Yanyan Zhao
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Andrew Shinkle
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Ilya Strunilin
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Cody Weimholt
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Kazuhito Sato
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Lijun Yao
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Mamatha Serasanambati
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Xiaolu Yang
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Matthew Wyczalkowski
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Houxiang Zhu
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Daniel Cui Zhou
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Reyka G Jayasinghe
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Daniel Mendez
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Michael C Wendl
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - David Clark
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21231, USA
| | | | - Yijun Ruan
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Melissa A Reimers
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Russell K Pachynski
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chris Kinsinger
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Scott Jewell
- Van Andel Institutes, Grand Rapids, MI, 49503, USA
| | - Daniel W Chan
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Aadel A Chaudhuri
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Milan G Chheda
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Benjamin D Humphreys
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - James J Hsieh
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Li Ding
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA.
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| | - Feng Chen
- Oncology Division, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| |
Collapse
|
65
|
Quah HS, Cao EY, Suteja L, Li CH, Leong HS, Chong FT, Gupta S, Arcinas C, Ouyang JF, Ang V, Celhar T, Zhao Y, Tay HC, Chan J, Takahashi T, Tan DSW, Biswas SK, Rackham OJL, Iyer NG. Single cell analysis in head and neck cancer reveals potential immune evasion mechanisms during early metastasis. Nat Commun 2023; 14:1680. [PMID: 36973261 PMCID: PMC10042873 DOI: 10.1038/s41467-023-37379-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
Profiling tumors at single-cell resolution provides an opportunity to understand complexities underpinning lymph-node metastases in head and neck squamous-cell carcinoma. Single-cell RNAseq (scRNAseq) analysis of cancer-cell trajectories identifies a subpopulation of pre-metastatic cells, driven by actionable pathways including AXL and AURK. Blocking these two proteins blunts tumor invasion in patient-derived cultures. Furthermore, scRNAseq analyses of tumor-infiltrating CD8 + T-lymphocytes show two distinct trajectories to T-cell dysfunction, corroborated by their clonal architecture based on single-cell T-cell receptor sequencing. By determining key modulators of these trajectories, followed by validation using external datasets and functional experiments, we uncover a role for SOX4 in mediating T-cell exhaustion. Finally, interactome analyses between pre-metastatic tumor cells and CD8 + T-lymphocytes uncover a putative role for the Midkine pathway in immune-modulation and this is confirmed by scRNAseq of tumors from humanized mice. Aside from specific findings, this study demonstrates the importance of tumor heterogeneity analyses in identifying key vulnerabilities during early metastasis.
Collapse
Affiliation(s)
- Hong Sheng Quah
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
- Academic Clinical Program in Oncology, Duke-NUS Medical School, Singapore, Singapore
| | - Elaine Yiqun Cao
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Lisda Suteja
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Constance H Li
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
- Academic Clinical Program in Oncology, Duke-NUS Medical School, Singapore, Singapore
| | - Hui Sun Leong
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
| | - Fui Teen Chong
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
| | - Shilpi Gupta
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Camille Arcinas
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
- Academic Clinical Program in Oncology, Duke-NUS Medical School, Singapore, Singapore
| | - John F Ouyang
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Vivian Ang
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Teja Celhar
- HuNIT platform, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yunqian Zhao
- HuNIT platform, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hui Chen Tay
- HuNIT platform, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jerry Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Takeshi Takahashi
- Laboratory Animal Research Department, Central Institute for Experimental Animals (CIEA), Kawasaki, Japan
| | - Daniel S W Tan
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
- Academic Clinical Program in Oncology, Duke-NUS Medical School, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Subhra K Biswas
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Owen J L Rackham
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
- The Alan Turing Institute, The British Library, London, United Kingdom
| | - N Gopalakrishna Iyer
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore.
- Academic Clinical Program in Oncology, Duke-NUS Medical School, Singapore, Singapore.
- Department of Head and Neck Surgery, National Cancer Centre, Singapore, Singapore.
- Division of Medical Sciences, National Cancer Centre, Singapore, Singapore.
| |
Collapse
|
66
|
Tomasik B, Bieńkowski M, Górska Z, Gutowska K, Kumięga P, Jassem J, Duchnowska R. Molecular aspects of brain metastases in breast cancer. Cancer Treat Rev 2023; 114:102521. [PMID: 36736124 DOI: 10.1016/j.ctrv.2023.102521] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/29/2023]
Abstract
Brain metastases (BM) are a common and devastating manifestation of breast cancer (BC). BM are particularly frequent in the HER2-positive and triple-negative breast cancer phenotypes and usually occur following the metastatic spread to extracranial sites. Several genes mediating BM and biomarkers predicting their risk in BC have been reported in the past decade. These findings have advanced the understanding of BM pathobiology and paved the way for developing new therapeutic strategies but they still warrant a thorough clinical validation. Hence, a better understanding of the mechanistic aspects of BM and delineating the interactions of tumor cells with the brain microenvironment are of utmost importance. This review discusses the molecular basis of the metastatic cascade: the epithelial-mesenchymal transition, cancer, and tumor microenvironment interaction and intravasation, priming of the metastatic niche in the brain, and survival in the new site. We also outline the postulated mechanisms of BC cells' brain tropism. Finally, we discuss advances in the field of biomarkers (both tissue-based and liquid-based) that predict BM from BC.
Collapse
Affiliation(s)
- Bartłomiej Tomasik
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, 17 Smoluchowskiego St., 80-214 Gdansk, Poland.
| | - Michał Bieńkowski
- Department of Pathology, Medical University of Gdańsk, 17 Smoluchowskiego St., 80-214 Gdańsk, Poland.
| | - Zuzanna Górska
- Department of Oncology, Military Institute of Medicine, 128 Szaserów St., 04-141 Warsaw, Poland.
| | - Klaudia Gutowska
- Department of Internal Diseases and Endocrinology, Medical University of Warsaw, 02-091 Warsaw, Poland; Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland.
| | - Paulina Kumięga
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland.
| | - Jacek Jassem
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, 17 Smoluchowskiego St., 80-214 Gdansk, Poland.
| | - Renata Duchnowska
- Department of Oncology, Military Institute of Medicine, 128 Szaserów St., 04-141 Warsaw, Poland.
| |
Collapse
|
67
|
Minikes AM, Song Y, Feng Y, Yoon C, Yoon SS, Jiang X. E-cadherin is a biomarker for ferroptosis sensitivity in diffuse gastric cancer. Oncogene 2023; 42:848-857. [PMID: 36717701 PMCID: PMC10291936 DOI: 10.1038/s41388-023-02599-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/31/2023]
Abstract
Gastric cancer is the third most common cause of cancer-related death worldwide. Diffuse-type gastric cancer (DGC) is a particularly aggressive subtype that is both difficult to detect and treat. DGC is distinguished by weak cell-cell cohesion, most often due to loss of the cell adhesion protein E-cadherin, a common occurrence in highly invasive, metastatic cancer cells. In this study, we demonstrate that loss-of-function mutation of E-cadherin in DGC cells results in their increased sensitivity to the non-apoptotic, iron-dependent form of cell death, ferroptosis. Homophilic contacts between E-cadherin molecules on adjacent cells suppress ferroptosis through activation of the Hippo pathway. Furthermore, single nucleotide mutations observed in DGC patients that ablate the homophilic binding capacity of E-cadherin reverse the ability of E-cadherin to suppress ferroptosis in both cell culture and xenograft models. Importantly, although E-cadherin loss in cancer cells is considered an essential event for epithelial-mesenchymal transition and subsequent metastasis, we found that circulating DGC cells lacking E-cadherin expression possess lower metastatic ability, due to their increased susceptibility to ferroptosis. Together, this study suggests that E-cadherin is a biomarker predicting the sensitivity to ferroptosis of DGC cells, both in primary tumor tissue and in circulation, thus guiding the usage of future ferroptosis-inducing therapeutics for the treatment of DGC.
Collapse
Affiliation(s)
- Alexander M Minikes
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yu Song
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Department of Oncology, Affiliated Zhangjiagang Hospital, Soochow University, Suzhou, China.
| | - Yan Feng
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Changhwan Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sam S Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
68
|
PELI1 and EGFR cooperate to promote breast cancer metastasis. Oncogenesis 2023; 12:9. [PMID: 36841821 PMCID: PMC9968314 DOI: 10.1038/s41389-023-00457-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/27/2023] Open
Abstract
Pellino-1 (PELI1) is an E3 ubiquitin ligase acting as a key regulator for the inflammation and autoimmunity via the ubiquitination of the substrate proteins. There is increasing evidence to support that PELI1 functions as an oncoprotein in tumorigenesis and metastasis. However, the molecular mechanism underlying the high expression and oncogenic roles of PELI1 in cancers remains limited. Herein, we revealed a novel regulation mechanism by which PELI1 and EGFR cooperate to promote breast cancer metastasis. EGFR is positively correlated with PELI1 expression in breast cancers, and its activation led to the phosphorylation of PELI1 at Tyr154 and Thr264, which subsequently activated its E3 ubiquitin ligase. Simultaneously, PELI1 physically interacted with and enhanced the stability of EGFR via the K63-linked polyubiquitination in reverse. The co-inhibition of the PELI1-EGFR showed synergetic effect to repress breast cancer metastasis. Furthermore, we identified a compound S62 as a small molecule disruptor of PELI1/EGFR that effectively repressed breast cancer metastasis. Our study not only uncovered the emerging roles of PELI1/EGFR interaction in the progression of breast cancer, but also provided an effective strategy for the inhibition of metastasis in breast cancer.
Collapse
|
69
|
Li D, Xia L, Huang P, Wang Z, Guo Q, Huang C, Leng W, Qin S. Heterogeneity and plasticity of epithelial-mesenchymal transition (EMT) in cancer metastasis: Focusing on partial EMT and regulatory mechanisms. Cell Prolif 2023:e13423. [PMID: 36808651 DOI: 10.1111/cpr.13423] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/05/2023] [Accepted: 01/27/2023] [Indexed: 02/22/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) or mesenchymal-epithelial transition (MET) plays critical roles in cancer metastasis. Recent studies, especially those based on single-cell sequencing, have revealed that EMT is not a binary process, but a heterogeneous and dynamic disposition with intermediary or partial EMT states. Multiple double-negative feedback loops involved by EMT-related transcription factors (EMT-TFs) have been identified. These feedback loops between EMT drivers and MET drivers finely regulate the EMT transition state of the cell. In this review, the general characteristics, biomarkers and molecular mechanisms of different EMT transition states were summarized. We additionally discussed the direct and indirect roles of EMT transition state in tumour metastasis. More importantly, this article provides direct evidence that the heterogeneity of EMT is closely related to the poor prognosis in gastric cancer. Notably, a seesaw model was proposed to explain how tumour cells regulate themselves to remain in specific EMT transition states, including epithelial state, hybrid/intermediate state and mesenchymal state. Additionally, this article also provides a review of the current status, limitations and future perspectives of EMT signalling in clinical applications.
Collapse
Affiliation(s)
- Dandan Li
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Pan Huang
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Zidi Wang
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Qiwei Guo
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Congcong Huang
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
70
|
Lin YP, Hseu YC, Thiyagarajan V, Vadivalagan C, Pandey S, Lin KY, Hsu YT, Liao JW, Lee CC, Yang HL. The in vitro and in vivo anticancer activities of Antrodia salmonea through inhibition of metastasis and induction of ROS-mediated apoptotic and autophagic cell death in human glioblastoma cells. Biomed Pharmacother 2023; 158:114178. [PMID: 36916401 DOI: 10.1016/j.biopha.2022.114178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Antrodia salmonea (AS) exhibits anticancer activities against various cancers. OBJECTIVE This study investigated the anticancer activities of AS on human glioblastoma (GBM8401 and U87MG) cells both in vitro and in vivo and explained the underlying molecular mechanism. METHODS MTT, colony formation, migration/invasion assay, immunoblotting, immunofluorescence, TUNEL, Annexin V/PI staining, AO staining, GFP-LC3 transfection, TEM, qPCR, siLC3, DCFH2-DA assay, and xenografted-nude mice were used to assess the potential of AS therapy. RESULTS AS treatment retarded growth and suppressed colony formation in glioblastoma cells. AS attenuates EMT by suppressing invasion and migration, increasing E-cadherin expression, decreasing Twist, Snail, and N-cadherin expression, and inhibiting Wnt/β-catenin pathways in GBM8401 and U87MG cells. Furthermore, AS induced apoptosis by activating caspase-3, cleaving PARP, and dysregulating Bax and Bcl-2 in both cell lines. TUNEL assay and Annexin V/PI staining indicated AS-mediated late apoptosis. Interestingly, AS induced autophagic cell death by LC3-II accumulation, AVO formation, autophagosome GFP-LC3 puncta, p62/SQSTM1 expression, and ATG4B inhibition in GBM8401 and U87MG cells. TEM data revealed that AS favored autophagosome and autolysosome formation. The autophagy inhibitors 3-MA/CQ and LC3 knockdown suppressed AS-induced apoptosis in glioblastoma cells, indicating that the inhibition of autophagy decreased AS-induced apoptosis. Notably, the antioxidant N-acetylcysteine (NAC) inhibited AS-mediated ROS production and AS-induced apoptotic and autophagic cell death. Furthermore, AS induced ROS-mediated inhibition of the PI3K/AKT/mTOR signaling pathway. AS reduced the tumor burden in GBM8401-xenografted nude mice and significantly modulated tumor xenografts by inducing anti-EMT, apoptosis, and autophagy. AS could be a potential antitumor agent in human glioblastoma treatment.
Collapse
Affiliation(s)
- Yi-Pin Lin
- Institute of Nutrition, College of Health Care, China Medical University, Taichung 40402, Taiwan, ROC
| | - You-Cheng Hseu
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung 40402, Taiwan, ROC; Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan, ROC; Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan, ROC; Research Center of Chinese Herbal Medicine, China Medical University, Taichung 40402, Taiwan, ROC.
| | - Varadharajan Thiyagarajan
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung 40402, Taiwan, ROC
| | - Chithravel Vadivalagan
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung 40402, Taiwan, ROC
| | - Sudhir Pandey
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Kai-Yuan Lin
- Department of Medical Research, Chi-Mei Medical Center, Tainan 710, Taiwan, ROC; Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan, ROC
| | - Yuan-Tai Hsu
- Institute of Nutrition, College of Health Care, China Medical University, Taichung 40402, Taiwan, ROC
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung-Hsing University, Taichung 402, Taiwan, ROC
| | - Chuan-Chen Lee
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan, ROC
| | - Hsin-Ling Yang
- Institute of Nutrition, College of Health Care, China Medical University, Taichung 40402, Taiwan, ROC.
| |
Collapse
|
71
|
Luo D, Zeng X, Zhang S, Li D, Cheng Z, Wang Y, Long J, Hu Z, Long S, Zhou J, Zhang S, Zeng Z. Pirfenidone suppressed triple-negative breast cancer metastasis by inhibiting the activity of the TGF-β/SMAD pathway. J Cell Mol Med 2023; 27:456-469. [PMID: 36651490 PMCID: PMC9889661 DOI: 10.1111/jcmm.17673] [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: 08/31/2022] [Revised: 12/20/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Among breast cancer patients, metastases are the leading cause of death. Despite decades of effort, little progress has been made to improve the treatment of breast cancer metastases, especially triple-negative breast cancer (TNBC). The extracellular matrix plays an important role in tumour growth and metastasis by causing its deposition, remodelling, and signalling. As we know, the process of fibrosis results in excessive amounts of extracellular matrix being deposited within the cells. So, it will be interesting to study if the use of anti-fibrotic drugs in combination with conventional chemotherapy drugs can produce synergistic antitumor effects. In this study, we assessed the efficacy of Pirfenidone (PFD), an FDA-approved medication for the treatment of idiopathic pulmonary fibrosis, on TNBC cells as well as its anti-tumour effects in xenograft tumour model. PFD inhibited in a dose-dependent manner breast cancer cell proliferation, migration, and invasion, while promoted their apoptosis in vitro. PFD also suppressed TGF-β-induced activation of Smad signalling pathway and expression level of EMT-inducing transcription factors (e.g. SNAI2, TWIST1, ZEB1) as well as the mesenchymal genes such as VIMENTIN and N-Cadherin. On the contrary, the expression level of epithelial marker gene E-Cadherin was up-regulated in the presence of PFD. In vivo, PFD alone exerted a milder but significant anti-tumour effect than the chemotherapy drug nanoparticle albumin-bound paclitaxel (nab-PTX) did in the breast cancer xenograft mouse model. Interestingly, PFD synergistically boosted the cancer-killing effect of nab-PTX. Furthermore, Our data suggest that PFD suppressed breast cancer metastasis by inhibiting the activity of the TGFβ/SMAD pathway.
Collapse
Affiliation(s)
- Daiqin Luo
- School of Basic Medical Sciences/School of Biology & EngineeringGuizhou Medical UniversityGuiyangChina,Engineering Center of cellular immunotherapy of Guizhou ProvinceGuiyangChina,Department of oncologyAffiliated Hospital of Guizhou Medical UniversityGuiyangChina,Department of oncologyAffiliated Cancer Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xianlin Zeng
- School of Basic Medical Sciences/School of Biology & EngineeringGuizhou Medical UniversityGuiyangChina,Engineering Center of cellular immunotherapy of Guizhou ProvinceGuiyangChina,Key Laboratory of infectious immunity and antibody engineering of Guizhou ProvinceGuiyangChina
| | - Shuling Zhang
- School of Public HealthGuizhou Medical UniversityGuiyangChina
| | - Daohong Li
- School of Basic Medical Sciences/School of Biology & EngineeringGuizhou Medical UniversityGuiyangChina,Engineering Center of cellular immunotherapy of Guizhou ProvinceGuiyangChina,Key Laboratory of infectious immunity and antibody engineering of Guizhou ProvinceGuiyangChina
| | - Zhimei Cheng
- Department of Interventional RadiologyAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Yun Wang
- School of Basic Medical Sciences/School of Biology & EngineeringGuizhou Medical UniversityGuiyangChina,Engineering Center of cellular immunotherapy of Guizhou ProvinceGuiyangChina,Key Laboratory of infectious immunity and antibody engineering of Guizhou ProvinceGuiyangChina,Key Laboratory of Endemic and Ethnic Diseases, Ministry of EducationGuizhou Medical UniversityGuiyangChina,State Key Laboratory of Functions & Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
| | - Jinhua Long
- School of Basic Medical Sciences/School of Biology & EngineeringGuizhou Medical UniversityGuiyangChina,Department of oncologyAffiliated Hospital of Guizhou Medical UniversityGuiyangChina,Department of oncologyAffiliated Cancer Hospital of Guizhou Medical UniversityGuiyangChina
| | - Zuquan Hu
- School of Basic Medical Sciences/School of Biology & EngineeringGuizhou Medical UniversityGuiyangChina,Engineering Center of cellular immunotherapy of Guizhou ProvinceGuiyangChina,Key Laboratory of infectious immunity and antibody engineering of Guizhou ProvinceGuiyangChina,Key Laboratory of Endemic and Ethnic Diseases, Ministry of EducationGuizhou Medical UniversityGuiyangChina,State Key Laboratory of Functions & Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
| | - Shiqi Long
- School of Basic Medical Sciences/School of Biology & EngineeringGuizhou Medical UniversityGuiyangChina,Engineering Center of cellular immunotherapy of Guizhou ProvinceGuiyangChina,Key Laboratory of infectious immunity and antibody engineering of Guizhou ProvinceGuiyangChina,Key Laboratory of Endemic and Ethnic Diseases, Ministry of EducationGuizhou Medical UniversityGuiyangChina,State Key Laboratory of Functions & Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
| | - Jing Zhou
- School of Basic Medical Sciences/School of Biology & EngineeringGuizhou Medical UniversityGuiyangChina,Engineering Center of cellular immunotherapy of Guizhou ProvinceGuiyangChina,Key Laboratory of infectious immunity and antibody engineering of Guizhou ProvinceGuiyangChina,Key Laboratory of Endemic and Ethnic Diseases, Ministry of EducationGuizhou Medical UniversityGuiyangChina,State Key Laboratory of Functions & Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
| | - Shuai Zhang
- Department of Interventional RadiologyAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Zhu Zeng
- School of Basic Medical Sciences/School of Biology & EngineeringGuizhou Medical UniversityGuiyangChina,Engineering Center of cellular immunotherapy of Guizhou ProvinceGuiyangChina,Key Laboratory of infectious immunity and antibody engineering of Guizhou ProvinceGuiyangChina,Key Laboratory of Endemic and Ethnic Diseases, Ministry of EducationGuizhou Medical UniversityGuiyangChina,State Key Laboratory of Functions & Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
72
|
Remigante A, Spinelli S, Marino A, Pusch M, Morabito R, Dossena S. Oxidative Stress and Immune Response in Melanoma: Ion Channels as Targets of Therapy. Int J Mol Sci 2023; 24:ijms24010887. [PMID: 36614330 PMCID: PMC9821408 DOI: 10.3390/ijms24010887] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Oxidative stress and immune response play an important role in the development of several cancers, including melanoma. Ion channels are aberrantly expressed in tumour cells and regulate neoplastic transformation, malignant progression, and resistance to therapy. Ion channels are localized in the plasma membrane or other cellular membranes and are targets of oxidative stress, which is particularly elevated in melanoma. At the same time, ion channels are crucial for normal and cancer cell physiology and are subject to multiple layers of regulation, and therefore represent promising targets for therapeutic intervention. In this review, we analyzed the effects of oxidative stress on ion channels on a molecular and cellular level and in the context of melanoma progression and immune evasion. The possible role of ion channels as targets of alternative therapeutic strategies in melanoma was discussed.
Collapse
Affiliation(s)
- Alessia Remigante
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy
| | - Sara Spinelli
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy
| | - Angela Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy
| | - Michael Pusch
- Biophysics Institute, National Research Council, 16149 Genova, Italy
| | - Rossana Morabito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy
- Correspondence:
| | - Silvia Dossena
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020 Salzburg, Austria
| |
Collapse
|
73
|
Zhong W, Sun T. Editorial: Epithelial-mesenchymal transition (EMT) as a therapeutic target in cancer. Front Oncol 2023; 13:1121416. [PMID: 36890820 PMCID: PMC9987418 DOI: 10.3389/fonc.2023.1121416] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Affiliation(s)
- Weilong Zhong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin Medical University, Tianjin, China
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| |
Collapse
|
74
|
Jayaraman H, Anandhapadman A, Ghone NV. In Vitro and In Vivo Comparative Analysis of Differentially Expressed Genes and Signaling Pathways in Breast Cancer Cells on Interaction with Mesenchymal Stem Cells. Appl Biochem Biotechnol 2023; 195:401-431. [PMID: 36087230 DOI: 10.1007/s12010-022-04119-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2022] [Indexed: 01/13/2023]
Abstract
The interaction of breast cancer cells (BCC) with mesenchymal stem cells (MSC) plays a vital role in influencing the gene expression in breast cancer cells and thereby its uncontrolled proliferation, metastasis, and drug resistance. The extent of MSC governing the BCC or the extent of BCC influencing the MSC is a complex process, and the interaction strongly depends upon conditions such as the presence or absence of other cell types and in vivo tumor microenvironment or simple in vitro conditions. Hence, understanding this interaction through gene expression profiling may provide key insights about potential genes which can be targeted for breast cancer treatment. In the current study, in vitro microarray dataset and in vivo RNA-seq dataset of BCC on interaction with the MSC were downloaded from NCBI GEO database and analyzed for differentially expressed genes (DEGs), gene ontology (GO) term enrichment, and Reactome pathway analysis. To target the genes which have similar effect on both in vitro and in vivo, a comparative analysis was performed, 24 genes were commonly upregulated in both in vitro and in vivo datasets, while no common downregulated genes were observed. Out of which, 16 significant genes based upon fold change (logFC > 2) are identified for manipulating the interactions between MSC and BCC. Among them, 6 of the identified genes (FSTL1, LOX, SERPINE1, INHBA, FN1, and VEGFA) have already been reported to be upregulated in BCC on interaction with MSC by various studies. Further experiments need to be conducted to understand the role of remaining 10 identified genes (EFEMP1, IGFBP3, EDIL3, IFITM1, IGFBP4, ITGA5, SLC3A2, HRH1, PPP1R15A, and NNMT) in MSC-BCC interaction. In addition to the reported significant genes and its associated pathways, the expression of long non-coding RNA identified in this study may increase our understanding about the way MSC interacts with BCC and accelerate MSC-based treatment strategies for breast cancer.
Collapse
Affiliation(s)
- Hariharan Jayaraman
- Department of Biotechnology, Sri Venkateswara College of Engineering, Post Bag No. 1, Sriperumbudur Taluk, 602117, Kancheepuram, Tamil Nadu, India
| | - Ashwin Anandhapadman
- Department of Biotechnology, Sri Venkateswara College of Engineering, Post Bag No. 1, Sriperumbudur Taluk, 602117, Kancheepuram, Tamil Nadu, India
| | - Nalinkanth Veerabadran Ghone
- Department of Chemical Engineering, Sri Sivasubramaniya Nadar College of Engineering, Rajiv Gandhi Salai (OMR), Kalavakkam, 603110, Tamil Nadu, India.
| |
Collapse
|
75
|
Chan MKK, Chan ELY, Ji ZZ, Chan ASW, Li C, Leung KT, To KF, Tang PMK. Transforming growth factor-β signaling: from tumor microenvironment to anticancer therapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:316-343. [PMID: 37205317 PMCID: PMC10185444 DOI: 10.37349/etat.2023.00137] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/09/2023] [Indexed: 05/21/2023] Open
Abstract
Transforming growth factor-β (TGF-β) signaling is an important pathway for promoting the pathogenesis of inflammatory diseases, including cancer. The roles of TGF-β signaling are heterogeneous and versatile in cancer development and progression, both anticancer and protumoral actions are reported. Interestingly, increasing evidence suggests that TGF-β enhances disease progression and drug resistance via immune-modulatory actions in the tumor microenvironment (TME) of solid tumors. A better understanding of its regulatory mechanisms in the TME at the molecular level can facilitate the development of precision medicine to block the protumoral actions of TGF-β in the TME. Here, the latest information about the regulatory mechanisms and translational research of TGF-β signaling in the TME for therapeutic development had been summarized.
Collapse
Affiliation(s)
- Max Kam-Kwan Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Emily Lok-Yiu Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Zoey Zeyuan Ji
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Alex Siu-Wing Chan
- Department of Applied Social Sciences, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Chunjie Li
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Kam-Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
- Correspondence: Patrick Ming-Kuen Tang, Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|
76
|
Mohseni Garakani M, Cooke ME, Weber MH, Wertheimer MR, Ajji A, Rosenzweig DH. A 3D, Compartmental Tumor-Stromal Microenvironment Model of Patient-Derived Bone Metastasis. Int J Mol Sci 2022; 24:ijms24010160. [PMID: 36613604 PMCID: PMC9820116 DOI: 10.3390/ijms24010160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Bone is a frequent site of tumor metastasis. The bone-tumor microenvironment is heterogeneous and complex in nature. Such complexity is compounded by relations between metastatic and bone cells influencing their sensitivity/resistance to chemotherapeutics. Standard chemotherapeutics may not show efficacy for every patient, and new therapeutics are slow to emerge, owing to the limitations of existing 2D/3D models. We previously developed a 3D interface model for personalized therapeutic screening, consisting of an electrospun poly lactic acid mesh activated with plasma species and seeded with stromal cells. Tumor cells embedded in an alginate-gelatin hydrogel are overlaid to create a physiologic 3D interface. Here, we applied our 3D model as a migration assay tool to verify the migratory behavior of different patient-derived bone metastasized cells. We assessed the impact of two different chemotherapeutics, Doxorubicin and Cisplatin, on migration of patient cells and their immortalized cell line counterparts. We observed different migratory behaviors and cellular metabolic activities blocked with both Doxorubicin and Cisplatin treatment; however, higher efficiency or lower IC50 was observed with Doxorubicin. Gene expression analysis of MDA-MB231 that migrated through our 3D hybrid model verified epithelial-mesenchymal transition through increased expression of mesenchymal markers involved in the metastasis process. Our findings indicate that we can model tumor migration in vivo, in line with different cell characteristics and it may be a suitable drug screening tool for personalized medicine approaches in metastatic cancer treatment.
Collapse
Affiliation(s)
- Mansoureh Mohseni Garakani
- Chemical Engineering Department, Polytechnique Montreal, Montreal, QC H3T1J4, Canada
- Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC H3T1J4, Canada
| | - Megan E. Cooke
- Department of Surgery, Division of Orthopaedic Surgery, McGill University, Montreal, QC H3G 1A4, Canada
- Injury, Repair and Recovery Program, Research Institute of McGill University Health Center (RI-MUHC), Montreal, QC H3G 1A4, Canada
| | - Michael H. Weber
- Department of Surgery, Division of Orthopaedic Surgery, McGill University, Montreal, QC H3G 1A4, Canada
- Injury, Repair and Recovery Program, Research Institute of McGill University Health Center (RI-MUHC), Montreal, QC H3G 1A4, Canada
| | - Michael R. Wertheimer
- Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC H3T1J4, Canada
- Department of Engineering Physics, Polytechnique Montreal, Montreal, QC H3T1J4, Canada
| | - Abdellah Ajji
- Chemical Engineering Department, Polytechnique Montreal, Montreal, QC H3T1J4, Canada
- Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC H3T1J4, Canada
- Correspondence: (A.A.); (D.H.R.); Tel.: +1-514-934-1934 (ext. 43238) (D.H.R.)
| | - Derek H. Rosenzweig
- Department of Surgery, Division of Orthopaedic Surgery, McGill University, Montreal, QC H3G 1A4, Canada
- Injury, Repair and Recovery Program, Research Institute of McGill University Health Center (RI-MUHC), Montreal, QC H3G 1A4, Canada
- Correspondence: (A.A.); (D.H.R.); Tel.: +1-514-934-1934 (ext. 43238) (D.H.R.)
| |
Collapse
|
77
|
Lu T, Zheng C, Fan Z. Cardamonin suppressed the migration, invasion, epithelial mesenchymal transition (EMT) and lung metastasis of colorectal cancer cells by down-regulating ADRB2 expression. PHARMACEUTICAL BIOLOGY 2022; 60:1011-1021. [PMID: 35645356 PMCID: PMC9154753 DOI: 10.1080/13880209.2022.2069823] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 03/09/2022] [Accepted: 04/11/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Cardamonin (CDN) can suppress cell growth in colorectal cancer (CRC), a common digestive malignancy. OBJECTIVE We explored the effect and mechanism of CDN on metastatic CRC. MATERIALS AND METHODS Two cell lines (HT29 and HCT116) were initially treated with CDN at different concentrations (5, 10 and 20 μmol/L) or 50 μmol/L propranolol (positive control) for 24 or 48 h. Then, the two cell lines were separately transfected with siADRB2 and ADRB2 overexpression plasmids, and further treated with 10 μmol/L CDN for 24 h. The cell viability, migration and invasion were determined by cell counting kit-8 (CCK-8), wound healing and transwell assays, respectively. The levels of ADRB2, matrix metalloprotease (MMP)-2, MMP-9, E-cadherin and N-cadherin were measured by Western blotting or/and RT-qPCR. A CRC metastasis model was established to evaluate the antimetastatic potential of CDN (25 mg/kg). RESULTS ADRB2 (3.2-fold change; p < 0.001) was highly expressed in CRC tissues. CDN at 10 μmol/L suppressed viability (69% and 70%), migration (33% and 66%), invasion (43% and 72%) and ADRB2 expression (2.2- and 2.84-fold change) in HT29 and HCT116 cells (p < 0.001). CDN at 10 μmol/L inhibited MMP-2, MMP-9 and N-cadherin expression but promoted E-cadherin expression in CRC cells (p < 0.001). Importantly, the effect of CDN on CRC cells was impaired by ADRB2 overexpression, but further enhanced by ADRB2 down-regulation (p < 0.01). Additionally, ADRB2 overexpression reversed the inhibitory effect of CDN on metastatic lung nodules (p < 0.05). Discussion and conclusions: CDN is a potential candidate for the treatment of metastatic CRC in clinical practice.
Collapse
Affiliation(s)
- Ting Lu
- Proctology Department, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunju Zheng
- Proctology Department, Huai’an TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Huai’an, China
| | - Zhimin Fan
- Proctology Department, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
78
|
Pore AA, Bithi SS, Zeinali M, Navaid HB, Nagrath S, Layeequr Rahman R, Vanapalli SA. Phenotyping of rare circulating cells in the blood of non-metastatic breast cancer patients using microfluidic Labyrinth technology. BIOMICROFLUIDICS 2022; 16:064107. [PMID: 36536791 PMCID: PMC9759355 DOI: 10.1063/5.0129602] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/30/2022] [Indexed: 05/13/2023]
Abstract
Label-free technologies for isolating rare circulating cells in breast cancer patients are widely available; however, they are mostly validated on metastatic patient blood samples. Given the need to use blood-based biomarkers to inform on disease progression and treatment decisions, it is important to validate these technologies in non-metastatic patient blood samples. In this study, we specifically focus on a recently established label-free microfluidic technology Labyrinth and assess its capabilities to phenotype a variety of rare circulating tumor cells indicative of epithelial-to-mesenchymal transition as well as cancer-associated macrophage-like (CAML) cells. We specifically chose a patient cohort that is non-metastatic and selected to undergo neoadjuvant chemotherapy to assess the performance of the Labyrinth technology. We enrolled 21 treatment naïve non-metastatic breast cancer patients of various disease stages. Our results indicate that (i) Labyrinth microfluidic technology is successfully able to isolate different phenotypes of CTCs despite the counts being low. (ii) Invasive phenotypes of CTCs such as transitioning CTCs and mesenchymal CTCs were found to be present in high numbers in stage III patients as compared to stage II patients. (iii) As the total load of CTCs increased, the mesenchymal CTCs were found to be increasing. (iv) Labyrinth was able to isolate CAMLs with the counts being higher in stage III patients as compared to stage II patients. Our study demonstrates the ability of the Labyrinth microfluidic technology to isolate rare cancer-associated cells from the blood of treatment naïve non-metastatic breast cancer patients, laying the foundation for tracking oncogenic spread and immune response in patients undergoing neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Adity A. Pore
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas 79409, USA
| | - Swastika S. Bithi
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas 79409, USA
| | - Mina Zeinali
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 79430, USA
| | - Hunaiz Bin Navaid
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas 79409, USA
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 79430, USA
| | | | - Siva A. Vanapalli
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas 79409, USA
| |
Collapse
|
79
|
Dukel M. Combination of naringenin and epicatechin sensitizes colon carcinoma cells to anoikis via regulation of the epithelial–mesenchymal transition (EMT). Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00317-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
80
|
Mukherjee B, Tiwari A, Palo A, Pattnaik N, Samantara S, Dixit M. Reduced expression of FRG1 facilitates breast cancer progression via GM-CSF/MEK-ERK axis by abating FRG1 mediated transcriptional repression of GM-CSF. Cell Death Dis 2022; 8:442. [PMID: 36329016 PMCID: PMC9633810 DOI: 10.1038/s41420-022-01240-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Multiple molecular subtypes and distinct clinical outcomes in breast cancer, necessitate specific therapy. Moreover, despite the improvements in breast cancer therapy, it remains the fifth cause of cancer-related deaths, indicating the involvement of unknown genes. To identify novel contributors and molecular subtype independent therapeutic options, we report reduced expression of FRG1 in breast cancer patients, which regulates GM-CSF expression via direct binding to its promoter. Reduction in FRG1 expression enhanced EMT and increased cell proliferation, migration, and invasion, in breast cancer cell lines. Loss of FRG1 increased GM-CSF levels which activated MEK/ERK axis and prevented apoptosis by inhibiting p53 in an ERK-dependent manner. FRG1 depletion in the mouse model increased tumor volume, phospho-ERK, and EMT marker levels. The therapeutic potential of anti-GM-CSF therapy was evident by reduced tumor size, when tumors with decreased FRG1 were treated with anti-GM-CSF mAb. We found an inverse expression pattern of FRG1 and phospho-ERK levels in breast cancer patient tissues, corroborating the in vitro and mouse model-based findings. Our findings first time elucidate the role of FRG1 as a metastatic suppressor of breast cancer by regulating the GM-CSF/MEK-ERK axis.
Collapse
Affiliation(s)
- Bratati Mukherjee
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, 752050, India.,Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, 400094, India
| | - Ankit Tiwari
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, 752050, India.,Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, 400094, India
| | - Ananya Palo
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, 752050, India.,Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, 400094, India
| | | | - Subrat Samantara
- Acharya Harihar Regional Cancer Centre (AHRCC), Cuttack, 753007, Odisha, India
| | - Manjusha Dixit
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, 752050, India. .,Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, 400094, India.
| |
Collapse
|
81
|
Liu P, Wang Z, Ou X, Wu P, Zhang Y, Wu S, Xiao X, Li Y, Ye F, Tang H. The FUS/circEZH2/KLF5/ feedback loop contributes to CXCR4-induced liver metastasis of breast cancer by enhancing epithelial-mesenchymal transition. Mol Cancer 2022; 21:198. [PMID: 36224562 PMCID: PMC9555172 DOI: 10.1186/s12943-022-01653-2] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/01/2022] [Indexed: 12/24/2022] Open
Abstract
Background Metastasis of breast cancer have caused the majority of cancer-related death worldwide. The circRNAs are associated with tumorigenesis and metastasis in breast cancer according to recent research. However, the biological mechanism of circRNAs in liver metastatic breast cancer remains ambiguous yet. Methods Microarray analysis of three pairs of primary BC tissues and matched hepatic metastatic specimens identified circEZH2. We used RT-qPCR and FISH assays to confirm circEZH2 existence, characteristics, and expression. Both in vivo and in vitro, circEZH2 played an oncogenic role which promoted metastasis as well. A range of bioinformatic analysis, Western blot, RNA pull-down, RIP, ChIP, and animal experiments were used to define the feedback loop involving FUS, circEZH2, miR-217-5p, KLF5, FUS, CXCR4 as well as epithelial and mesenchymal transition. Results In our research, circEZH2 was proved to be upregulated in liver metastases in BC and predicted the worse prognosis in breast cancer patients. Overexpression of circEZH2 notably accentuated the vitality and invasion of BC cells, whereas knockdown of circEZH2 elicited the literally opposite effects. Besides, overexpressed circEZH2 promoted tumorigenesis and liver metastasis in vivo. Moreover, circEZH2 could adsorb miR-217-5p to upregulate KLF5 thus leading to activate FUS transcription which would facilitate the back-splicing program of circEZH2. Meanwhile, KLF5 could upregulated CXCR4 transcriptionally to accelerate epithelial and mesenchymal transition of breast cancer. Conclusions Consequently, a novel feedback loop FUS/circEZH2/KLF5/CXCR4 was established while circEZH2 could be novel biomarker and potential target for BC patients’ therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-022-01653-2.
Collapse
Affiliation(s)
- Peng Liu
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zehao Wang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xueqi Ou
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Peng Wu
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yue Zhang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Song Wu
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xiangsheng Xiao
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yuehua Li
- Department of Medical Oncology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China.
| | - Feng Ye
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Hailin Tang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
82
|
AVA-NP-695 Selectively Inhibits ENPP1 to Activate STING Pathway and Abrogate Tumor Metastasis in 4T1 Breast Cancer Syngeneic Mouse Model. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196721. [PMID: 36235254 PMCID: PMC9573294 DOI: 10.3390/molecules27196721] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/24/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
Cyclic GMP-AMP synthase (cGAS) is an endogenous DNA sensor that synthesizes cyclic guanosine monophosphate–adenosine monophosphate (2′3′-cGAMP) from ATP and GTP. 2′3′-cGAMP activates the stimulator of interferon genes (STING) pathway, resulting in the production of interferons and pro-inflammatory cytokines. Ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) is the phosphodiesterase that negatively regulates the STING pathway by hydrolyzing 2′3′-cGAMP. It has been established that the cGAS–STING pathway plays a major role in inhibiting tumor growth by upregulating T cell response. Herein, we demonstrate that AVA-NP-695, a selective and highly potent ENPP1 inhibitor, apart from the immunomodulatory effect also modulates cancer metastasis by negatively regulating epithelial–mesenchymal transition (EMT). We established that the combined addition of 2′3′-cGAMP and AVA-NP-695 significantly abrogated the transforming growth factor beta (TGF-ꞵ)-induced EMT in MDA-MB-231 cells. Finally, results from the in vivo study showed superior tumor growth inhibition and impact on tumor metastasis of AVA-NP-695 compared to Olaparib and PD-1 in a syngeneic 4T1 breast cancer mouse model. The translation of efficacy from in vitro to in vivo 4T1 tumor model provides a strong rationale for the therapeutic potential of AVA-NP-695 against triple-negative breast cancer (TNBC) as an immunomodulatory and anti-metastatic agent.
Collapse
|
83
|
Five EMT-Related Gene Signatures Predict Acute Myeloid Leukemia Patient Outcome. DISEASE MARKERS 2022; 2022:7826393. [PMID: 36246561 PMCID: PMC9568336 DOI: 10.1155/2022/7826393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/07/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022]
Abstract
Background The epithelial mesenchymal transition (EMT) gene has been shown to be significantly associated with the prognosis of solid tumors; however, there is a lack of models for the EMT gene to predict the prognosis of AML patients. Methods First, we downloaded clinical data and raw transcriptome sequencing data from the TCGA database of acute myeloid leukemia (AML) patients. All currently confirmed EMT-related genes were obtained from the dbEMT 2.0 database, and 30% of the TCGA data were randomly selected as the test set. Univariate Cox regression analysis, random forest, and lasso regression were used to optimize the number of genes for model construction, and multivariate Cox regression was used for model construction. Area under the ROC curve was used to assess the efficacy of the model application, and the internal validation set was used to assess the stability of the model. Results A total of 173 AML samples were downloaded, and a total of 1184 EMT-related genes were downloaded. The results of univariate batch Cox regression analysis suggested that 212 genes were associated with patient prognosis, random forest and lasso regression yielded 18 and 8 prognosis-related EMT genes, respectively, and the results of multifactorial COX regression model suggested that 5 genes, CBR1, HS3ST3B1, LIMA1, MIR573, and PTP4A3, were considered as independent risk factors affecting patient prognosis. The model ROC results suggested that the area under the curve was 0.868 and the internal validation results showed that the area under the curve was 0.815. Conclusion During this study, we constructed a signature model of five EMT-related genes to predict overall survival in patients with AML; it will provide a useful tool for clinical decision making.
Collapse
|
84
|
Devanaboyina M, Kaur J, Whiteley E, Lin L, Einloth K, Morand S, Stanbery L, Hamouda D, Nemunaitis J. NF-κB Signaling in Tumor Pathways Focusing on Breast and Ovarian Cancer. Oncol Rev 2022; 16:10568. [PMID: 36531159 PMCID: PMC9756851 DOI: 10.3389/or.2022.10568] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/02/2022] [Indexed: 08/30/2023] Open
Abstract
Immune disorders and cancer share a common pathway involving NF-κb signaling. Through involvement with GM-CSF, NF-κB can contribute to proliferation and activation of T- and B- cells as well as immune cell migration to sites of inflammation. In breast cancer, this signaling pathway has been linked to resistance with endocrine and chemotherapies. Similarly, in ovarian cancer, NF-κB influences angiogenesis and inflammation pathways. Further, BRCA1 signaling common to both breast and ovarian cancer also has the capability to induce NF-κB activity. Immunotherapy involving NF-κB can also be implemented to combat chemoresistance. The complex signaling pathways of NF-κB can be harnessed for developing cancer therapeutics to promote immunotherapy for improving patient outcomes.
Collapse
Affiliation(s)
- Monika Devanaboyina
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Jasskiran Kaur
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Emma Whiteley
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Leslie Lin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Katelyn Einloth
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Susan Morand
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | | | - Danae Hamouda
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | | |
Collapse
|
85
|
Li Y, Hung SW, Zhang R, Man GCW, Zhang T, Chung JPW, Fang L, Wang CC. Melatonin in Endometriosis: Mechanistic Understanding and Clinical Insight. Nutrients 2022; 14:nu14194087. [PMID: 36235740 PMCID: PMC9572886 DOI: 10.3390/nu14194087] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Endometriosis is defined as the development of endometrial glands and stroma outside the uterine cavity. Pathophysiology of this disease includes abnormal hormone profiles, cell survival, migration, invasion, angiogenesis, oxidative stress, immunology, and inflammation. Melatonin is a neuroendocrine hormone that is synthesized and released primarily at night from the mammalian pineal gland. Increasing evidence has revealed that melatonin can be synthesized and secreted from multiple extra-pineal tissues where it regulates immune response, inflammation, and angiogenesis locally. Melatonin receptors are expressed in the uterus, and the therapeutic effects of melatonin on endometriosis and other reproductive disorders have been reported. In this review, key information related to the metabolism of melatonin and its biological effects is summarized. Furthermore, the latest in vitro and in vivo findings are highlighted to evaluate the pleiotropic functions of melatonin, as well as to summarize its physiological and pathological effects and treatment potential in endometriosis. Moreover, the pharmacological and therapeutic benefits derived from the administration of exogenous melatonin on reproductive system-related disease are discussed to support the potential of melatonin supplements toward the development of endometriosis. More clinical trials are needed to confirm its therapeutic effects and safety.
Collapse
Affiliation(s)
- Yiran Li
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Sze-Wan Hung
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Ruizhe Zhang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Gene Chi-Wai Man
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Tao Zhang
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Jacqueline Pui-Wah Chung
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Lanlan Fang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Correspondence: (L.F.); (C.-C.W.); Tel.: +86-371-6691-3635 (L.F.); +852-3505-4267 (C.-C.W.)
| | - Chi-Chiu Wang
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Hong Kong 999077, China
- Laboratory of Reproduction and Development, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Chinese University of Hong Kong-Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
- Correspondence: (L.F.); (C.-C.W.); Tel.: +86-371-6691-3635 (L.F.); +852-3505-4267 (C.-C.W.)
| |
Collapse
|
86
|
Roshanazadeh MR, Adelipour M, Sanaei A, Chenane H, Rashidi M. TRIM3 and TRIM16 as potential tumor suppressors in breast cancer patients. BMC Res Notes 2022; 15:312. [PMID: 36180926 PMCID: PMC9523982 DOI: 10.1186/s13104-022-06193-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/07/2022] [Indexed: 11/10/2022] Open
Abstract
Objective Breast cancer is the leading cause of death among women in many countries. Numerous factors serve as oncogenes or tumor suppressors in breast cancer. The large family of Tripartite-motif (TRIM) proteins with ~ 80 members has drawn attention for their role in cancer. TRIM3 and TRIM16 have shown suppressive activity in different cancers. This study aimed to evaluate the expression of TRIM3 and TRIM16 in cancerous and normal breast samples and to investigate their association with different clinical and pathological parameters. Results qRT-PCR was utilized to determine the gene expression of TRIM3 and TRIM16. The expression of TRIM3 and TRIM16 genes in tumor samples were significantly reduced to 0.45 and 0.29 fold, respectively. TRIM3 and TRIM16 genes expression were both positively correlated with the invasion of breast cancer. TRIM3 gene expression was associated with tumors’ histological grade. However, no significant association was found between the expression of the genes and tumor size, stage and necrosis. The expression of TRIM3 and TRIM16 are significantly reduced in breast cancer tissues. Besides, the expression of both TRIM3 and TRIM16 genes significantly plummet in lymphatic/vascular and perineural invasive samples. Hence, we suggest a potential tumor suppressor role for TRIM3 and TRIM16 in breast cancer.
Collapse
Affiliation(s)
- Mohammad Reza Roshanazadeh
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of clinical biochemistry, Faculty of medicine, jundishapour University of medical sciences, Ahvaz, Iran
| | - Maryam Adelipour
- Department of clinical biochemistry, Faculty of medicine, jundishapour University of medical sciences, Ahvaz, Iran
| | - Arash Sanaei
- Department of clinical biochemistry, Faculty of medicine, jundishapour University of medical sciences, Ahvaz, Iran
| | - Hadi Chenane
- Department of clinical biochemistry, Faculty of medicine, jundishapour University of medical sciences, Ahvaz, Iran
| | - Mojtaba Rashidi
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. .,Department of clinical biochemistry, Faculty of medicine, jundishapour University of medical sciences, Ahvaz, Iran.
| |
Collapse
|
87
|
Zhang DD, Sun XL, Liang ZY, Wang XY, Zhang LN. FAM96A and FAM96B function as new tumor suppressor genes in breast cancer through regulation of the Wnt/β-catenin signaling pathway. Life Sci 2022; 308:120983. [PMID: 36165859 DOI: 10.1016/j.lfs.2022.120983] [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: 07/13/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/26/2022]
Abstract
AIMS Family with sequence similarity 96 member A and B (FAM96A and FAM96B) are two highly conserved homologous proteins belonging to MIP18 family. Some studies have shown that FAM96A and FAM96B are significantly down-regulated in human gastrointestinal stromal tumors, colon cancer, and liver cancer. However, the molecular mechanisms of FAM96A/B in breast cancer are unknown. This work aims to explore the roles of FAM96A/B in breast cancer progression. MAIN METHODS Specific siRNAs were used to down-regulate FAM96A/B expression, and recombinant plasmids were used to up-regulate FAM96A/B expression in breast cancer cells. Cell proliferation was measured using MTT and colony formation. Cell cycle and apoptosis were detected by flow cytometry. Cell migration and invasion were examined by wound healing and transwell assays. The relationships among FAM96A/B, EMT and Wnt/β-catenin pathway were determined by analyzing expression changes of classical markers. KEY FINDINGS We found that FAM96A/B expression was down-regulated in breast cancer. FAM96A/B overexpression suppressed breast cancer cell proliferation, invasion and migration, induced cell apoptosis and caused cell cycle arrest. Conversely, FAM96A/B knockdown exhibited the opposite effects. Moreover, our data demonstrated that FAM96A/B overexpression suppressed EMT and Wnt/β-catenin pathway, while FAM96A/B knockdown showed the promoting effects on EMT and Wnt/β-catenin pathway. Furthermore, a Wnt pathway inhibitor, XAV-939 reversed the promoting effects of FAM96A/B knockdown on breast cancer progression. SIGNIFICANCE Our findings suggest that FAM96A/B may function as new tumor suppressor genes and inhibit breast cancer progression via modulating Wnt/β-catenin pathway, which can provide the potential markers for breast cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Di-Di Zhang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Xiao-Lin Sun
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Zhao-Yuan Liang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Xin-Ya Wang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Li-Na Zhang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
88
|
Liu Z, Fan M, Xuan X, Xia C, Huang G, Ma L. Celastrol inhibits the migration and invasion and enhances the anti-cancer effects of docetaxel in human triple-negative breast cancer cells. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:189. [PMID: 36071249 DOI: 10.1007/s12032-022-01792-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 07/07/2022] [Indexed: 10/14/2022]
Abstract
The molecular mechanism of anti-metastatic effect of celastrol is not fully understood in breast cancer cells. Herein, we investigated the activity and molecular mechanism of celastrol in triple-negative breast cancer (TNBC) cells, which is a more aggressive subtype of breast cancer. The results of wound healing assay and trans-well assay revealed that celastrol inhibited cell migration and invasion under sub-cytotoxic concentrations in MDA-MB-231 and MDA-MB-468 TNBC cells. Molecular data showed that the effect of celastrol on TNBC cells might be mediated via up-regulation of E-cadherin, a key protein involved in epithelial-mesenchymal transition (EMT). In addition, Hakai, an E3 ligase responsible for E-cadherin complex ubiquitination and degradation, was down-regulated under celastrol treatment. Hakai partially contributed to celastrol-induced anti-invasive effect. In addition, celastrol and docetaxel could synergistically inhibit growth and metastasis of MDA-MB-231 cells. Our results showing anti-migratory/anti-invasive effects of celastrol and associated mechanisms provide new evidence for the development of celastrol as a potential anti-metastatic compound against highly aggressive breast cancer, and celastrol in combination with docetaxel might potentially be used as a novel regimen for the treatment of TNBC.
Collapse
Affiliation(s)
- Zi Liu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, People's Republic of China
| | - Minghui Fan
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, People's Republic of China
| | - Xiaojing Xuan
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, People's Republic of China
| | - Chenlu Xia
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, People's Republic of China
| | - Guozheng Huang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, People's Republic of China
| | - Liang Ma
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, 59 Hudong Road, Ma'anshan, 243002, Anhui, People's Republic of China.
| |
Collapse
|
89
|
Characterization of Vemurafenib-Resistant Melanoma Cell Lines Reveals Novel Hallmarks of Targeted Therapy Resistance. Int J Mol Sci 2022; 23:ijms23179910. [PMID: 36077308 PMCID: PMC9455970 DOI: 10.3390/ijms23179910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Regardless of the significant improvements in treatment of melanoma, the majority of patients develop resistance whose mechanisms are still not completely understood. Hence, we generated and characterized two melanoma-derived cell lines, primary WM793B and metastatic A375M, with acquired resistance to the RAF inhibitor vemurafenib. The morphology of the resistant primary WM793B melanoma cells showed EMT-like features and exhibited a hybrid phenotype with both epithelial and mesenchymal characteristics. Surprisingly, the vemurafenib-resistant melanoma cells showed a decreased migration ability but also displayed a tendency to collective migration. Signaling pathway analysis revealed the reactivation of MAPK and the activation of the PI3K/AKT pathway depending on the vemurafenib-resistant cell line. The acquired resistance to vemurafenib caused resistance to chemotherapy in primary WM793B melanoma cells. Furthermore, the cell-cycle analysis and altered levels of cell-cycle regulators revealed that resistant cells likely transiently enter into cell cycle arrest at the G0/G1 phase and gain slow-cycling cell features. A decreased level of NME1 and NME2 metastasis suppressor proteins were found in WM793B-resistant primary melanoma, which is possibly the result of vemurafenib-acquired resistance and is one of the causes of increased PI3K/AKT signaling. Further studies are needed to reveal the vemurafenib-dependent negative regulators of NME proteins, their role in PI3K/AKT signaling, and their influence on vemurafenib-resistant melanoma cell characteristics.
Collapse
|
90
|
Raeisi M, Zehtabi M, Velaei K, Fayyazpour P, Aghaei N, Mehdizadeh A. Anoikis in cancer: The role of lipid signaling. Cell Biol Int 2022; 46:1717-1728. [DOI: 10.1002/cbin.11896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 12/20/2022]
Affiliation(s)
- Mortaza Raeisi
- Hematology and Oncology Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Mojtaba Zehtabi
- Hematology and Oncology Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Kobra Velaei
- Department of Anatomical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Parisa Fayyazpour
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
| | - Negar Aghaei
- Department of Psycology, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
- Imam Sajjad Hospital Tabriz Azad University Tabriz Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
91
|
Ragab Ibrahim FA, Naser Hussein ZU, Yousef AI, Abd El Moneim NA, Hussein AM, Ahmed AFM, Ragab NM, Al-Masry O. Insights on possible interplay between epithelial-mesenchymal transition and T-type voltage gated calcium channels genes in metastatic breast carcinoma. Heliyon 2022; 8:e10160. [PMID: 36060991 PMCID: PMC9434037 DOI: 10.1016/j.heliyon.2022.e10160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 05/22/2022] [Accepted: 07/27/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) is the most common life-threatening malignancy amongst women with high incidence worldwide. In Egypt, it is the most known malignancy amongst females. Epithelial-mesenchymal transition (EMT) participates in breast tumors’ invasiveness, and metastasis, but the process is poorly understood. The involvement of voltage-gated calcium channels signaling in EMT has not yet been fully explored. Therefore, the aim of this study was to investigate the possible role of T-type calcium channels in metastasis and EMT among breast cancer patients. The study was carried out on 48 female breast cancer patients who were divided into two groups; metastatic and non-metastatic. qRT-PCR was employed to measure the expression of EMT marker genes (N- cadherin, E-cadherin, Snail, Vimentin and T-type VGCCs genes (CACNA1G, CACNA1H, and CACNA1I). The results of the present study revealed differential expression of the EMT marker genes in blood and tissue of non-metastatic and metastatic breast cancer patients, with a clear tendency for the mesenchymal markers to be significantly elevated in metastatic patients as well as malignant tissues taken from non-metastatic patients as compared to their paired tumor adjacent normal (TAN) tissue. Both CACNA1H and CACNA1I (T-type VGCCs oncogenes) were significantly elevated in blood of metastatic patients when compared to non-metastatic ones. In contrast, CACNA1G (tumor suppressor) exhibited a significant decrease in metastatic patients. The strong correlation between the expression of T-type VGCCs and mesenchymal marker genes in metastatic breast cancer patients casts light on the role of T-type VGCCs in metastasis and their involved in tumor invasiveness.
Collapse
|
92
|
Cai FF, Xu HR, Yu SH, Li P, Lu YY, Chen J, Bi ZQ, Sun HS, Cheng J, Zhuang HQ, Hua ZC. ADT-OH inhibits malignant melanoma metastasis in mice via suppressing CSE/CBS and FAK/Paxillin signaling pathway. Acta Pharmacol Sin 2022; 43:1829-1842. [PMID: 34795411 PMCID: PMC9253130 DOI: 10.1038/s41401-021-00799-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Hydrogen sulfide (H2S) is widely recognized as the third endogenous gas signaling molecule and may play a key role in cancer biological processes. ADT-OH (5-(4-hydroxyphenyl)-3H-1,2-dithiocyclopentene-3-thione) is one of the most widely used organic donors for the slow release of H2S and considered to be a potential anticancer compound. In this study, we investigated the antimetastatic effects of ADT-OH in highly metastatic melanoma cells. A tail-vein-metastasis model was established by injecting B16F10 and A375 cells into the tail veins of mice, whereas a mouse footpad-injection model was established by injecting B16F10 cells into mouse footpads. We showed that administration of ADT-OH significantly inhibited the migration and invasion of melanoma cells in the three different animal models. We further showed that ADT-OH dose-dependently inhibited the migration and invasion of B16F10, B16F1 and A375 melanoma cells as evaluated by wound healing and Transwell assays in vitro. LC-MS/MS and bioinformatics analyses revealed that ADT-OH treatment inhibited the EMT process in B16F10 and A375 cells by reducing the expression of FAK and the downstream response protein Paxillin. Overexpression of FAK reversed the inhibitory effects of ADT-OH on melanoma cell migration. Moreover, after ADT-OH treatment, melanoma cells showed abnormal expression of the H2S-producing enzymes CSE/CBS and the AKT signaling pathways. In addition, ADT-OH significantly suppressed the proliferation of melanoma cells. Collectively, these results demonstrate that ADT-OH inhibits the EMT process in melanoma cells by suppressing the CSE/CBS and FAK signaling pathways, thereby exerting its antimetastatic activity. ADT-OH may be used as an antimetastatic agent in the future.
Collapse
Affiliation(s)
- Fang-Fang Cai
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210008, China
- School of Biopharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Huang-Ru Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210008, China
| | - Shi-Hui Yu
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210008, China
| | - Ping Li
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210008, China
| | - Yan-Yan Lu
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210008, China
| | - Jia Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210008, China
| | - Zhi-Qian Bi
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210008, China
| | - Hui-Song Sun
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210008, China
| | - Jian Cheng
- Institute of Neuroscience, Soochow University, Suzhou, 215031, China.
| | - Hong-Qin Zhuang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210008, China.
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210008, China.
- School of Biopharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc., Changzhou, 213164, China.
| |
Collapse
|
93
|
He S, Ning Y, Ma F, Liu D, Jiang S, Deng S. IL-23 Inhibits Trophoblast Proliferation, Migration, and EMT via Activating p38 MAPK Signaling Pathway to Promote Recurrent Spontaneous Abortion. J Microbiol Biotechnol 2022; 32:792-799. [PMID: 35637168 PMCID: PMC9628909 DOI: 10.4014/jmb.2112.12056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 12/15/2022]
Abstract
As a vital problem in reproductive health, recurrent spontaneous abortion (RSA) affects about 1% of women. We performed this study with an aim to explore the molecular mechanism of interleukin-23 (IL-23) and find optimal or effective methods to improve RSA. First, ELISA was applied to evaluate the expressions of IL-23 and its receptor in HTR-8/SVneo cells after IL-23 treatment. CCK-8, TUNEL, wound healing and transwell assays were employed to assess the proliferation, apoptosis, migration and invasion of HTR-8/SVneo cells, respectively. Additionally, the expressions of apoptosis-, migration-, epithelial-mesenchymal transition- (EMT-) and p38 MAPK signaling pathway-related proteins were measured by western blotting. To further investigate the relationship between IL-23 and p38 MAPK signaling pathway, HTR-8/SVneo cells were treated for 1 h with p38 MAPK inhibitor SB239063, followed by a series of cellular experiments on proliferation, apoptosis, migration and invasion, as aforementioned. The results showed that IL-23 and its receptors were greatly elevated in IL-23-treated HTR-8/SVneo cells. Additionally, IL-23 demonstrated suppressive effects on the proliferation, apoptosis, migration, invasion and EMT of IL-23-treated HTR-8/SVneo cells. More importantly, the molecular mechanism of IL-23 was revealed in this study; that is to say, IL-23 inhibited the proliferation, apoptosis, migration, invasion and EMT of IL-23-treated HTR-8/SVneo cells via activating p38 MAPK signaling pathway. In conclusion, IL-23 inhibits trophoblast proliferation, migration, and EMT via activating p38 MAPK signaling pathway, suggesting that IL-23 might be a novel target for the improvement of RSA.
Collapse
Affiliation(s)
- Shan He
- Department of Pharmacy, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Yan Ning
- Department of Traditional Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China,Corresponding author Phone: +0755-82889999 E-mail:
| | - Fei Ma
- Department of Traditional Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Dayan Liu
- Department of Genesiology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Shaoyan Jiang
- Department of Pharmacy, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Shaojie Deng
- Department of Pharmacy, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| |
Collapse
|
94
|
Breast Cancer Metastasis: Mechanisms and Therapeutic Implications. Int J Mol Sci 2022; 23:ijms23126806. [PMID: 35743249 PMCID: PMC9224686 DOI: 10.3390/ijms23126806] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the most common malignancy in women worldwide. Metastasis is the leading cause of high mortality in most cancers. Although predicting the early stage of breast cancer before metastasis can increase the survival rate, breast cancer is often discovered or diagnosed after metastasis has occurred. In general, breast cancer has a poor prognosis because it starts as a local disease and can spread to lymph nodes or distant organs, contributing to a significant impediment in breast cancer treatment. Metastatic breast cancer cells acquire aggressive characteristics from the tumor microenvironment (TME) through several mechanisms including epithelial–mesenchymal transition (EMT) and epigenetic regulation. Therefore, understanding the nature and mechanism of breast cancer metastasis can facilitate the development of targeted therapeutics focused on metastasis. This review discusses the mechanisms leading to metastasis and the current therapies to improve the early diagnosis and prognosis in patients with metastatic breast cancer.
Collapse
|
95
|
Kang YH, Wang JH, Lee JS, Lee NH, Son CG. Coptidis Rhizoma Suppresses Metastatic Behavior by Inhibiting TGF-β-Mediated Epithelial-Mesenchymal Transition in 5-FU-Resistant HCT116 Cells. Front Pharmacol 2022; 13:909331. [PMID: 35770076 PMCID: PMC9234293 DOI: 10.3389/fphar.2022.909331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/31/2022] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the second most lethal malignancy worldwide. The high mortality rate of CRC is largely due to cancer metastasis. Recently, suppressing epithelial-to-mesenchymal transition (EMT) has been considered a promising strategy for treating metastatic cancer, especially drug-resistant metastatic cancer. The present study aimed to evaluate the antimetastatic effect of Coptidis Rhizoma, as well as the potential underlying mechanisms, using a 5-fluorouracil-resistant colon tumor cell model (HCT116/R). Coptidis Rhizoma 30% ethanol extract (CRE) significantly inhibited HCT116/R cells migration and invasion. CRE effectively inhibited EMT in HCT116/R cells by upregulating the expression of an epithelial marker (E-cadherin) and downregulating the expression of mesenchymal markers (vimentin, Snail, and ZEB2) at both the protein and gene levels. Immunofluorescence assays also confirmed consistent patterns in the levels of E-cadherin and vimentin. In addition, the anti-EMT activity of CRE and its related effects were associated with the CRE-mediated suppression of the TGF-β pathway, as shown by changes in the levels of downstream molecules (phosphorylated Akt and p38), and inhibition of migration, invasion, and protein expression of TGF-β after treatment/cotreatment with a TGF-β inhibitor (SB431542). In conclusion, Coptidis Rhizoma exerts an antimetastatic effect, especially in the treatment of drug-resistant cancer, and the possible mechanisms are associated with inhibiting EMT via TGF-β signaling. Thus, Coptidis Rhizoma will likely become a potential therapeutic candidate for simultaneously mitigating drug resistance and metastasis in CRC.
Collapse
Affiliation(s)
- Yong-Hwi Kang
- Institute of Bioscience and Integrative Medicine, Daejeon Oriental Hospital of Daejeon University, Daejeon, South Korea
| | - Jing-Hua Wang
- Institute of Bioscience and Integrative Medicine, Daejeon Oriental Hospital of Daejeon University, Daejeon, South Korea
| | - Jin-Seok Lee
- Institute of Bioscience and Integrative Medicine, Daejeon Oriental Hospital of Daejeon University, Daejeon, South Korea
| | - Nam-Hun Lee
- Institute of Bioscience and Integrative Medicine, Daejeon Oriental Hospital of Daejeon University, Daejeon, South Korea
- Department of Clinical Oncology, Cheonan Oriental Hospital of Daejeon University, Cheonan-si, South Korea
- *Correspondence: Nam-Hun Lee, ; Chang-Gue Son,
| | - Chang-Gue Son
- Institute of Bioscience and Integrative Medicine, Daejeon Oriental Hospital of Daejeon University, Daejeon, South Korea
- *Correspondence: Nam-Hun Lee, ; Chang-Gue Son,
| |
Collapse
|
96
|
Sirtuins and Hypoxia in EMT Control. Pharmaceuticals (Basel) 2022; 15:ph15060737. [PMID: 35745656 PMCID: PMC9228842 DOI: 10.3390/ph15060737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 05/25/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT), a physiological process during embryogenesis, can become pathological in the presence of different driving forces. Reduced oxygen tension or hypoxia is one of these forces, triggering a large number of molecular pathways with aberrant EMT induction, resulting in cancer and fibrosis onset. Both hypoxia-induced factors, HIF-1α and HIF-2α, act as master transcription factors implicated in EMT. On the other hand, hypoxia-dependent HIF-independent EMT has also been described. Recently, a new class of seven proteins with deacylase activity, called sirtuins, have been implicated in the control of both hypoxia responses, HIF-1α and HIF-2α activation, as well as EMT induction. Intriguingly, different sirtuins have different effects on hypoxia and EMT, acting as either activators or inhibitors, depending on the tissue and cell type. Interestingly, sirtuins and HIF can be activated or inhibited with natural or synthetic molecules. Moreover, recent studies have shown that these natural or synthetic molecules can be better conveyed using nanoparticles, representing a valid strategy for EMT modulation. The following review, by detailing the aspects listed above, summarizes the interplay between hypoxia, sirtuins, and EMT, as well as the possible strategies to modulate them by using a nanoparticle-based approach.
Collapse
|
97
|
Asadirad A, Khodadadi A, Talaiezadeh A, Shohan M, Rashno M, Joudaki N. Evaluation of miRNA-21-5p and miRNA-10b-5p levels in serum-derived exosomes of breast cancer patients in different grades. Mol Cell Probes 2022; 64:101831. [PMID: 35660458 DOI: 10.1016/j.mcp.2022.101831] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND & OBJECTIVES Tumor cells have various effects and dominance over other healthy cells. Cancer cells alter the cell program in healthy cells by secreting exosomes containing microRNAs involved in epithelial-mesenchymal transition (EMT). They can migrate to distant organs and establish a pre-metastatic niche. The purpose of this study was to determine the expression of miRNA-21-5p and miRNA-10b-5p, both of which are involved in EMT, in breast cancer-derived exosomes of various grades in order to identify new biomarkers involved in breast cancer progression. METHODS In this study, a blood sample was taken from 60 patients with grades I, II, or III breast cancer, as well as twenty healthy individuals as a control group. The exosomes were then purified from serum samples, and their relative expression of miRNA-21-5p and miRNA-10b-5p was determined using the real-time PCR method. RESULTS miRNA-21-5p expression was significantly increased in patients with breast cancer grades I, II, and III compared to the control group (p < 0.01), (p < 0.0001) and (p < 0.0001), respectively, as was miRNA-10b-5p expression in patients with breast cancer grades I, II, and III compared to the control group (p < 0.0001), (p < 0.0001) and (p < 0.0001), respectively. CONCLUSION Our results show that both microRNAs increase as cells lose their differentiation and become more invasive, which is evidence of cancer progression. Hence, both microRNAs may have the potential to be used alone or in combination with other biomarkers for the diagnosis and prognosis of breast cancer.
Collapse
Affiliation(s)
- Ali Asadirad
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Khodadadi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Abdolhassan Talaiezadeh
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Surgery, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Shohan
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Rashno
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nazanin Joudaki
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
98
|
Thymidylate Synthase Overexpression Drives the Invasive Phenotype in Colon Cancer Cells. Biomedicines 2022; 10:biomedicines10061267. [PMID: 35740289 PMCID: PMC9219882 DOI: 10.3390/biomedicines10061267] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/12/2022] Open
Abstract
Thymidylate synthase (TYMS) is the crucial enzymatic precursor for DNA biosynthesis and, therefore, the critical target for numerous types of chemotherapy, including the most frequently applied agent in colon cancer treatment 5-fluorouracil (5-FU). TYMS also seems to be associated with cancer metastasis and acquiring mesenchymal character by tumor cells during epithelial–mesenchymal transition (EMT). Based on that knowledge, we decided to investigate the role of TYMS in the modulation of invasive ability in colon cancer cells, where its effect on cancer metastasis has not been studied in detail before. We employed colon cancer cells isolated from different stages of tumor development, cells undergoing EMT, and TYMS overexpressing cells. The elongation ratio, cell migration, invasion assay, and MMP-7 secretion were applied to analyze the cell behavior. Important epithelial and mesenchymal markers characteristic of EMT were examined at the protein level by Western blot assay. Overall, our study showed a correlation between TYMS level and invasion ability in colon cancer cells and, above all, a crucial role of TYMS in the EMT regulation. We postulate that chemotherapeutics that decrease or inhibit TYMS expression could increase the effectiveness of the therapy in patients with colon cancer, especially in the metastatic stage.
Collapse
|
99
|
Xiao J, McGill JR, Nasir A, Lekan A, Johnson B, Wilkins DJ, Pearson GW, Tanner K, Goodarzi H, Glasgow E, Schlegel R, Agarwal S. Identifying drivers of breast cancer metastasis in progressively invasive subpopulations of zebrafish-xenografted MDA-MB-231. MOLECULAR BIOMEDICINE 2022; 3:16. [PMID: 35614362 PMCID: PMC9133282 DOI: 10.1186/s43556-022-00080-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/09/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer metastasis is the primary cause of the high mortality rate among human cancers. Efforts to identify therapeutic agents targeting cancer metastasis frequently fail to demonstrate efficacy in clinical trials despite strong preclinical evidence. Until recently, most preclinical studies used mouse models to evaluate anti-metastatic agents. Mouse models are time-consuming and expensive. In addition, an important drawback is that mouse models inadequately model the early stages of metastasis which plausibly leads to the poor correlation with clinical outcomes. Here, we report an in vivo model based on xenografted zebrafish embryos where we select for progressively invasive subpopulations of MDA-MB-231 breast cancer cells. A subpopulation analogous to circulating tumor cells found in human cancers was selected by injection of MDA-MB-231 cells into the yolk sacs of 2 days post-fertilized zebrafish embryos and selecting cells that migrated to the tail. The selected subpopulation derived from MDA-MB-231 cells were increasingly invasive in zebrafish. Isolation of these subpopulations and propagation in vitro revealed morphological changes consistent with activation of an epithelial-mesenchymal transition program. Differential gene analysis and knockdown of genes identified gene-candidates (DDIT4, MT1X, CTSD, and SERPINE1) as potential targets for anti-metastasis therapeutics. Furthermore, RNA-splicing analysis reinforced the importance of BIRC5 splice variants in breast cancer metastasis. This is the first report using zebrafish to isolate and expand progressively invasive populations of human cancer cells. The model has potential applications in understanding the metastatic process, identification and/or development of therapeutics that specifically target metastatic cells and formulating personalized treatment strategies for individual cancer patients.
Collapse
Affiliation(s)
- Jerry Xiao
- Department of Pathology, Center for Cell Reprogramming, Georgetown University, Washington, DC, USA.,Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Joseph R McGill
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Apsra Nasir
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Alexander Lekan
- Department of Pathology, Center for Cell Reprogramming, Georgetown University, Washington, DC, USA
| | - Bailey Johnson
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Devan J Wilkins
- Department of Pathology, Center for Cell Reprogramming, Georgetown University, Washington, DC, USA.,Eastern Virginia Medical School, Norfolk, VA, USA
| | - Gray W Pearson
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Kandice Tanner
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hani Goodarzi
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Eric Glasgow
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Richard Schlegel
- Department of Pathology, Center for Cell Reprogramming, Georgetown University, Washington, DC, USA
| | - Seema Agarwal
- Department of Pathology, Center for Cell Reprogramming, Georgetown University, Washington, DC, USA.
| |
Collapse
|
100
|
Faria CC, Cascão R, Custódia C, Paisana E, Carvalho T, Pereira P, Roque R, Pimentel J, Miguéns J, Cortes-Ciriano I, Barata JT. Patient-derived models of brain metastases recapitulate human disseminated disease. Cell Rep Med 2022; 3:100623. [PMID: 35584628 PMCID: PMC9133464 DOI: 10.1016/j.xcrm.2022.100623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 02/01/2022] [Accepted: 04/08/2022] [Indexed: 12/02/2022]
Abstract
Dissemination of cancer cells from primary tumors to the brain occurs in many cancer patients, increasing morbidity and death. There is an unmet medical need to develop translational platforms to evaluate therapeutic responses. Toward this goal, we established a library of 23 patient-derived xenografts (PDXs) of brain metastases (BMs) from eight distinct primary tumors. In vivo tumor formation correlates with patients’ poor survival. Mouse subcutaneous xenografts develop spontaneous metastases and intracardiac PDXs increase dissemination to the CNS, both models mimicking the dissemination pattern of the donor patient. We test the FDA-approved drugs buparlisib (pan-PI3K inhibitor) and everolimus (mTOR inhibitor) and show their efficacy in treating our models. Finally, we show by RNA sequencing that human BMs and their matched PDXs have similar transcriptional profiles. Overall, these models of BMs recapitulate the biology of human metastatic disease and can be valuable translational platforms for precision medicine. Established PDXs of brain metastasis from multiple cancers PDXs recapitulate the dissemination pattern of patient tumors Patient-derived models of brain metastases are valuable to test anticancer drugs Human brain metastases and their PDXs retain similar transcriptional profiles
Collapse
Affiliation(s)
- Claudia C Faria
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisboa, Portugal.
| | - Rita Cascão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos Custódia
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Eunice Paisana
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tânia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Pedro Pereira
- Laboratory of Neuropathology, Neurology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisboa, Portugal
| | - Rafael Roque
- Laboratory of Neuropathology, Neurology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisboa, Portugal
| | - José Pimentel
- Laboratory of Neuropathology, Neurology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisboa, Portugal
| | - José Miguéns
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisboa, Portugal
| | - Isidro Cortes-Ciriano
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD, UK
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|