1
|
Lan Y, Liu D, Liang B, Song X, Xie L, Peng H, Guo H, Hong C, Weng X, Wei X, Liao X, Liang R, Huang D, Liu M. ITGA3-MET interaction promotes papillary thyroid cancer progression via ERK and PI3K/AKT pathways. Ann Med 2025; 57:2483379. [PMID: 40138447 PMCID: PMC11948363 DOI: 10.1080/07853890.2025.2483379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 04/18/2024] [Accepted: 05/10/2024] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Studies have examined the role of integrin α3 (ITGA3) in papillary thyroid carcinoma (PTC). However, the functional and molecular mechanism by which ITGA3 is involved in the progression of PTC remains poorly understood. METHODS To investigate the role of ITGA3 in PTC, raw PTC transcriptome data underwent comprehensive bioinformatics analyses, including differential expression, co-expression network, and enrichment analyses. ITGA3 expression was validated via immunohistochemistry and western blotting in PTC tissues. Cell functional assays and xenograft models assessed PTC cell behaviour. The potential mechanisms of ITGA3 were elucidated using bioinformatics analyses, western blotting, co-immunoprecipitation, and immunofluorescence. Finally, integration of ITGA3 expression with clinical parameters enabled nomogram construction for precise prediction of cervical lymph node metastasis (CLNM) in PTC. RESULTS ITGA3 was upregulated in PTC and associated strongly with CLNM (79.5% vs. 53.84%, p = 0.016). ITGA3 expression enhanced PTC proliferation and migration in vitro and in vivo via cooperating with the MET protein tyrosine kinase, followed by phosphorylation of MET at Tyr1234/1235, and activation of ERK and PI3K/AKT signaling pathways. Furthermore, upregulation ITGA3 reduced phosphorylation at FAK-Tyr397 and Src-Tyr416 in PTC cells. Finally, a nomogram combining ITGA3 expression and clinical parameters for predicting CLNM was constructed and validated, achieving a ROC curve AUC of 0.719, suggesting potential application for PTC diagnosis. CONCLUSIONS ITGA3 promotes PTC cell proliferation and migration by cooperating with MET to activate MET-ERK and MET-PI3K-AKT signalling. ITGA3-MET cooperation may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Youmian Lan
- Department of Head and Neck, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, China
| | - Dongchen Liu
- Department of Head and Neck, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Bin Liang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, China
| | - Xuhong Song
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, China
| | - Lingzhu Xie
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, China
| | - Hanwei Peng
- Department of Head and Neck, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Haipeng Guo
- Department of Head and Neck, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Chaoqun Hong
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xuwu Weng
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xiaolong Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xiaoqi Liao
- Department of Head and Neck, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Rui Liang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, China
| | - Dongyang Huang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Muyuan Liu
- Department of Head and Neck, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
2
|
Valipour M, Zakeri Khatir Z, Ayati A, Hosseini A, Sheibani M, Irannejad H. Advances in the selective c-MET kinase inhibitors: Application of fused [5,6]-Bicyclic nitrogen-containing cores for anticancer drug design. Eur J Med Chem 2025; 284:117177. [PMID: 39724725 DOI: 10.1016/j.ejmech.2024.117177] [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: 09/26/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Over the past two decades, small molecules bearing [5,6]-bicyclic nitrogen-containing cores have emerged as one of the most extensively studied structures for the development of selective c-MET kinase inhibitors. Structure-activity relationship (SAR) studies have demonstrated that modifying these cores can significantly impact the biological properties of c-MET inhibitors, including safety/toxicity, potency, and metabolic stability. For example, although c-MET kinase inhibitors containing the [1,2,4]triazolo[4,3-b][1,2,4]triazine scaffold (core P) exhibit high inhibitory potency, they often face challenges due to metabolic stability defects. Alternatively, compounds containing [1,2,3]triazolo[4,5-b]pyrazine (core K) and [1,2,4]triazolo[4,3-b]pyridazine (core I) scaffolds demonstrate lower potency but improved metabolic stability, allowing some of them to progress into clinical trials and even be approved as novel anticancer drugs. Fortunately, X-ray crystallography studies have well elucidated key interactions between [5,6]-bicyclic nitrogen-containing cores and crucial amino acid residues within the c-MET active site. These insights emphasize the significance of π-π stacking interactions with Tyr1230 and hydrogen bonding with Asp1222, providing valuable guidance for the targeted design and optimization of selective c-MET kinase inhibitors. Following the identification/introduction of sixteen distinct [5,6]-bicyclic nitrogen-containing cores (cores A-P) utilized in the design of selective c-MET kinase inhibitors over the past two decades, this manuscript offers a comprehensive review of their roles in drug development of anticancer agents, and describes the various synthesis methods employed. The insights presented herein can serve as a resource for better structural optimization of c-MET kinase inhibitors in the future research.
Collapse
Affiliation(s)
- Mehdi Valipour
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Zahra Zakeri Khatir
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Adileh Ayati
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asieh Hosseini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sheibani
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hamid Irannejad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
3
|
Lengyel M, Molnár Á, Nagy T, Jdeed S, Garai I, Horváth Z, Uray IP. Zymogen granule protein 16B (ZG16B) is a druggable epigenetic target to modulate the mammary extracellular matrix. Cancer Sci 2025; 116:81-94. [PMID: 39489500 PMCID: PMC11711063 DOI: 10.1111/cas.16382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/19/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024] Open
Abstract
High tissue density of the mammary gland is considered a pro-tumorigenic factor, hence suppressing the stimuli that induce matrix buildup carries the potential for cancer interception. We found that in non-malignant mammary epithelial cells the combination of the chemopreventive agents bexarotene (Bex) and carvedilol (Carv) suppresses the zymogen granule protein 16B (ZG16B, PAUF) through an interaction of ARID1A with a proximal enhancer. Bex + Carv also reduced ZG16B levels in vivo in normal breast tissue and MDA-MB231 tumor xenografts. The relevance of ZG16B is underscored by ongoing clinical trials targeting ZG16B in pancreatic cancers, but its role in breast cancer development is unclear. In immortalized mammary epithelial cells, secreted recombinant ZG16B stimulated mitogenic kinase phosphorylation, detachment and mesenchymal characteristics, and promoted proliferation, motility and clonogenic growth. Highly concerted induction of specific laminin, collagen and integrin isoforms indicated a shift in matrix properties toward increased density and cell-matrix interactions. Exogenous ZG16B alone blocked Bex + Carv-mediated control of cell growth and migration, and antagonized Bex + Carv-induced gene programs regulating cell adhesion and migration. In breast cancer cells ZG16B induced colony formation and anchorage-independent growth, and stimulated migration in a PI3K/Akt-dependent manner. In contrast, Bex + Carv inhibited colony formation, reduced Ki67 levels, ZG16B expression and glucose uptake in MDA-MB231 xenografts. These data establish ZG16B as a druggable pro-tumorigenic target in breast cell transformation and suggest a key role of the matrisome network in rexinoid-dependent antitumor activity.
Collapse
Affiliation(s)
- Máté Lengyel
- Department of Clinical Oncology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- The Molecular Cell and Immune Biology Doctoral SchoolUniversity of DebrecenDebrecenHungary
| | - Ádám Molnár
- Department of Clinical Oncology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Tamás Nagy
- Department of Nuclear MedicineUniversity of DebrecenDebrecenHungary
| | - Sham Jdeed
- Department of Clinical Oncology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- The Molecular Cell and Immune Biology Doctoral SchoolUniversity of DebrecenDebrecenHungary
| | - Ildikó Garai
- Department of Nuclear MedicineUniversity of DebrecenDebrecenHungary
| | - Zsolt Horváth
- Center of OncoradiologyBács‐Kiskun County Teaching HospitalKecskemétHungary
| | - Iván P. Uray
- Department of Clinical Oncology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- The Molecular Cell and Immune Biology Doctoral SchoolUniversity of DebrecenDebrecenHungary
- Department of Biochemistry and Molecular BiologyUniversity of DebrecenDebrecenHungary
| |
Collapse
|
4
|
Gallo S, Folco CB, Crepaldi T. The MET Oncogene Network of Interacting Cell Surface Proteins. Int J Mol Sci 2024; 25:13692. [PMID: 39769452 PMCID: PMC11728269 DOI: 10.3390/ijms252413692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
The MET oncogene, encoding the hepatocyte growth factor (HGF) receptor, plays a key role in tumorigenesis, invasion, and resistance to therapy, yet its full biological functions and activation mechanisms remain incompletely understood. A feature of MET is its extensive interaction network, encompassing the following: (i) receptor tyrosine kinases (RTKs); (ii) co-receptors (e.g., CDCP1, Neuropilin1); (iii) adhesion molecules (e.g., integrins, tetraspanins); (iv) proteases (e.g., ADAM10); and (v) other receptors (e.g., CD44, plexins, GPCRs, and NMDAR). These interactions dynamically modulate MET's activation, signaling, intracellular trafficking, and degradation, enhancing its functional versatility and oncogenic potential. This review offers current knowledge on MET's partnerships, focusing on their functional impact on signaling output, therapeutic resistance, and cellular behavior. Finally, we evaluate emerging combination therapies targeting MET and its interactors, highlighting their potential to overcome resistance and improve clinical outcomes. By exploring the complex interplay within the MET network of interacting cell surface proteins, this review provides insights into advancing anti-cancer strategies and understanding the broader implications of RTK crosstalk in oncology.
Collapse
Affiliation(s)
- Simona Gallo
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| | - Consolata Beatrice Folco
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| | - Tiziana Crepaldi
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| |
Collapse
|
5
|
Pierre HC, Amrine CSM, Doyle MG, Salvi A, Raja HA, Chekan JR, Huntsman AC, Fuchs JR, Liu K, Burdette JE, Pearce CJ, Oberlies NH. Verticillins: fungal epipolythiodioxopiperazine alkaloids with chemotherapeutic potential. Nat Prod Rep 2024; 41:1327-1345. [PMID: 38629495 PMCID: PMC11409914 DOI: 10.1039/d3np00068k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
Covering: 1970 through June of 2023Verticillins are epipolythiodioxopiperazine (ETP) alkaloids, many of which possess potent, nanomolar-level cytotoxicity against a variety of cancer cell lines. Over the last decade, their in vivo activity and mode of action have been explored in detail. Notably, recent studies have indicated that these compounds may be selective inhibitors of histone methyltransferases (HMTases) that alter the epigenome and modify targets that play a crucial role in apoptosis, altering immune cell recognition, and generating reactive oxygen species. Verticillin A (1) was the first of 27 analogues reported from fungal cultures since 1970. Subsequent genome sequencing identified the biosynthetic gene cluster responsible for producing verticillins, allowing a putative pathway to be proposed. Further, molecular sequencing played a pivotal role in clarifying the taxonomic characterization of verticillin-producing fungi, suggesting that most producing strains belong to the genus Clonostachys (i.e., Bionectria), Bionectriaceae. Recent studies have explored the total synthesis of these molecules and the generation of analogues via both semisynthetic and precursor-directed biosynthetic approaches. In addition, nanoparticles have been used to deliver these molecules, which, like many natural products, possess challenging solubility profiles. This review summarizes over 50 years of chemical and biological research on this class of fungal metabolites and offers insights and suggestions on future opportunities to push these compounds into pre-clinical and clinical development.
Collapse
Affiliation(s)
- Herma C Pierre
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, P.O. Box 26170, Greensboro, North Carolina 27402, USA.
| | - Chiraz Soumia M Amrine
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, P.O. Box 26170, Greensboro, North Carolina 27402, USA.
- Department of Physical and Earth Sciences. Arkansas Tech University, 1701 N. Boulder Ave., Russellville, Arkansas 72801, USA
| | - Michael G Doyle
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, P.O. Box 26170, Greensboro, North Carolina 27402, USA.
| | - Amrita Salvi
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 900 S. Ashland Ave (M/C 870), Chicago, Illinois 60607, USA
| | - Huzefa A Raja
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, P.O. Box 26170, Greensboro, North Carolina 27402, USA.
| | - Jonathan R Chekan
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, P.O. Box 26170, Greensboro, North Carolina 27402, USA.
| | - Andrew C Huntsman
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Ohio State University, 500 W. 12th Ave., Columbus, Ohio 43210, USA
| | - James R Fuchs
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Ohio State University, 500 W. 12th Ave., Columbus, Ohio 43210, USA
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology and the Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 900 S. Ashland Ave (M/C 870), Chicago, Illinois 60607, USA
| | | | - Nicholas H Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, P.O. Box 26170, Greensboro, North Carolina 27402, USA.
| |
Collapse
|
6
|
Lin C, Yang H, Luo Q, Liu Q. FAK mediates hypoxia-induced pulmonary artery smooth muscle cell proliferation by modulating mitochondrial transcription termination factor 1/cyclin D1. Clin Transl Sci 2024; 17:e13767. [PMID: 38488492 PMCID: PMC10941516 DOI: 10.1111/cts.13767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/18/2024] Open
Abstract
This study aimed to investigate the mechanism of FAK-dependent hypoxia-induced proliferation on human pulmonary artery smooth muscle cells (HPASMCs). Primary HPASMCs were isolated and cultured in vitro under normal and hypoxia conditions to assess cell proliferation with cell counting kit-8. FAK and mitochondrial transcription termination factor 1 (mTERF1) were silenced with siRNA, mRNA, and protein levels of FAK, mTERF1, and cyclin D1 were determined. HPASMC proliferation increased under hypoxia compared to normal conditions. Knocking down FAK or mTERF1 with siRNA led to decreased cell proliferation under both normal and hypoxia conditions. FAK knockdown led to the reduction of both mTERF1 and cyclin D1 expressions under the hypoxia conditions, whereas mTERF1 knockdown led to the downregulation of cyclin D1 expression but not FAK expression under the same condition. However, under normal conditions, knocking down either FAK or mTERF1 had no impact on cyclin D1 expression. These results suggested that FAK may regulate the mTERF1/cyclin D1 signaling pathway to modulate cell proliferation in hypoxia.
Collapse
Affiliation(s)
- Chunlong Lin
- Department of Respiratory and Critical Care MedicineYueyang People's Hospital of Hunan Normal UniversityYueyangChina
| | - Hui Yang
- Department of Respiratory and Critical Care MedicineYueyang People's Hospital of Hunan Normal UniversityYueyangChina
| | - Qiong Luo
- Department of Respiratory and Critical Care MedicineYueyang People's Hospital of Hunan Normal UniversityYueyangChina
| | - Qi Liu
- Department of Respiratory and Critical Care MedicineYueyang People's Hospital of Hunan Normal UniversityYueyangChina
| |
Collapse
|
7
|
Saeed A, Park R, Pathak H, Al-Bzour AN, Dai J, Phadnis M, Al-Rajabi R, Kasi A, Baranda J, Sun W, Williamson S, Chiu YC, Osmanbeyoglu HU, Madan R, Abushukair H, Mulvaney K, Godwin AK, Saeed A. Clinical and biomarker results from a phase II trial of combined cabozantinib and durvalumab in patients with chemotherapy-refractory colorectal cancer (CRC): CAMILLA CRC cohort. Nat Commun 2024; 15:1533. [PMID: 38378868 PMCID: PMC10879200 DOI: 10.1038/s41467-024-45960-2] [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: 10/17/2023] [Accepted: 01/31/2024] [Indexed: 02/22/2024] Open
Abstract
CAMILLA is a basket trial (NCT03539822) evaluating cabozantinib plus the ICI durvalumab in chemorefractory gastrointestinal cancer. Herein, are the phase II colorectal cohort results. 29 patients were evaluable. 100% had confirmed pMMR/MSS tumors. Primary endpoint was met with ORR of 27.6% (95% CI 12.7-47.2%). Secondary endpoints of 4-month PFS rate was 44.83% (95% CI 26.5-64.3%); and median OS was 9.1 months (95% CI 5.8-20.2). Grade≥3 TRAE occurred in 39%. In post-hoc analysis of patients with RAS wild type tumors, ORR was 50% and median PFS and OS were 6.3 and 21.5 months respectively. Exploratory spatial transcriptomic profiling of pretreatment tumors showed upregulation of VEGF and MET signaling, increased extracellular matrix activity and preexisting anti-tumor immune responses coexisting with immune suppressive features like T cell migration barriers in responders versus non-responders. Cabozantinib plus durvalumab demonstrated anti-tumor activity, manageable toxicity, and have led to the activation of the phase III STELLAR-303 trial.
Collapse
Affiliation(s)
- Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Robin Park
- Division of Hematology and Medical Oncology, Moffitt Cancer Cente, Tampa, FL, USA
| | - Harsh Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ayah Nedal Al-Bzour
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Junqiang Dai
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Milind Phadnis
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Raed Al-Rajabi
- Department of Medicine, Division of Medical Oncology, University of Kansas Medical Center, Kansas City, Ks, USA
- University of Kansas Cancer Center, Kansas City, KS, USA
| | - Anup Kasi
- Department of Medicine, Division of Medical Oncology, University of Kansas Medical Center, Kansas City, Ks, USA
- University of Kansas Cancer Center, Kansas City, KS, USA
| | - Joaquina Baranda
- Department of Medicine, Division of Medical Oncology, University of Kansas Medical Center, Kansas City, Ks, USA
- University of Kansas Cancer Center, Kansas City, KS, USA
| | - Weijing Sun
- Department of Medicine, Division of Medical Oncology, University of Kansas Medical Center, Kansas City, Ks, USA
- University of Kansas Cancer Center, Kansas City, KS, USA
| | - Stephen Williamson
- Department of Medicine, Division of Medical Oncology, University of Kansas Medical Center, Kansas City, Ks, USA
- University of Kansas Cancer Center, Kansas City, KS, USA
| | | | | | - Rashna Madan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Hassan Abushukair
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Kelly Mulvaney
- University of Kansas Cancer Center, Kansas City, KS, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Cancer Center, Kansas City, KS, USA
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Azhar Saeed
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT, USA
| |
Collapse
|
8
|
Modvig S, Jeyakumar J, Marquart HV, Christensen C. Integrins and the Metastasis-like Dissemination of Acute Lymphoblastic Leukemia to the Central Nervous System. Cancers (Basel) 2023; 15:cancers15092504. [PMID: 37173970 PMCID: PMC10177281 DOI: 10.3390/cancers15092504] [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: 03/31/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) disseminates with high prevalence to the central nervous system (CNS) in a process resembling aspects of the CNS surveillance of normal immune cells as well as aspects of brain metastasis from solid cancers. Importantly, inside the CNS, the ALL blasts are typically confined within the cerebrospinal fluid (CSF)-filled cavities of the subarachnoid space, which they use as a sanctuary protected from both chemotherapy and immune cells. At present, high cumulative doses of intrathecal chemotherapy are administered to patients, but this is associated with neurotoxicity and CNS relapse still occurs. Thus, it is imperative to identify markers and novel therapy targets specific to CNS ALL. Integrins represent a family of adhesion molecules involved in cell-cell and cell-matrix interactions, implicated in the adhesion and migration of metastatic cancer cells, normal immune cells, and leukemic blasts. The ability of integrins to also facilitate cell-adhesion mediated drug resistance, combined with recent discoveries of integrin-dependent routes of leukemic cells into the CNS, have sparked a renewed interest in integrins as markers and therapeutic targets in CNS leukemia. Here, we review the roles of integrins in CNS surveillance by normal lymphocytes, dissemination to the CNS by ALL cells, and brain metastasis from solid cancers. Furthermore, we discuss whether ALL dissemination to the CNS abides by known hallmarks of metastasis, and the potential roles of integrins in this context.
Collapse
Affiliation(s)
- Signe Modvig
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jenani Jeyakumar
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Hanne Vibeke Marquart
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Claus Christensen
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| |
Collapse
|
9
|
Rivas S, Marín A, Samtani S, González-Feliú E, Armisén R. MET Signaling Pathways, Resistance Mechanisms, and Opportunities for Target Therapies. Int J Mol Sci 2022; 23:ijms232213898. [PMID: 36430388 PMCID: PMC9697723 DOI: 10.3390/ijms232213898] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
The MET gene, known as MET proto-oncogene receptor tyrosine kinase, was first identified to induce tumor cell migration, invasion, and proliferation/survival through canonical RAS-CDC42-PAK-Rho kinase, RAS-MAPK, PI3K-AKT-mTOR, and β-catenin signaling pathways, and its driver mutations, such as MET gene amplification (METamp) and the exon 14 skipping alterations (METex14), activate cell transformation, cancer progression, and worse patient prognosis, principally in lung cancer through the overactivation of their own oncogenic and MET parallel signaling pathways. Because of this, MET driver alterations have become of interest in lung adenocarcinomas since the FDA approval of target therapies for METamp and METex14 in 2020. However, after using MET target therapies, tumor cells develop adaptative changes, favoring tumor resistance to drugs, the main current challenge to precision medicine. Here, we review a link between the resistance mechanism and MET signaling pathways, which is not only limited to MET. The resistance impacts MET parallel tyrosine kinase receptors and signals shared hubs. Therefore, this information could be relevant in the patient's mutational profile evaluation before the first target therapy prescription and follow-up to reduce the risk of drug resistance. However, to develop a resistance mechanism to a MET inhibitor, patients must have access to the drugs. For instance, none of the FDA approved MET inhibitors are registered as such in Chile and other developing countries. Constant cross-feeding between basic and clinical research will thus be required to meet future challenges imposed by the acquired resistance to targeted therapies.
Collapse
Affiliation(s)
- Solange Rivas
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7550000, Chile
| | - Arnaldo Marín
- Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| | - Suraj Samtani
- Departamento de Oncología Médica, Clínica Las Condes, Santiago 7550000, Chile
- Hospital Félix Bulnes, Santiago 9080000, Chile
| | - Evelin González-Feliú
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7550000, Chile
| | - Ricardo Armisén
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7550000, Chile
- Correspondence:
| |
Collapse
|
10
|
Acevedo DS, Fang WB, Rao V, Penmetcha V, Leyva H, Acosta G, Cote P, Brodine R, Swerdlow R, Tan L, Lorenzi PL, Cheng N. Regulation of growth, invasion and metabolism of breast ductal carcinoma through CCL2/CCR2 signaling interactions with MET receptor tyrosine kinases. Neoplasia 2022; 28:100791. [PMID: 35405500 PMCID: PMC9010752 DOI: 10.1016/j.neo.2022.100791] [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: 01/05/2022] [Revised: 03/02/2022] [Accepted: 03/21/2022] [Indexed: 12/26/2022]
Abstract
CCR2 correlates with MET receptor expression in breast ductal carcinomas. CCL2/CCR2 signaling in breast cancer cells depend on interactions with MET. CCR2 and MET signals alter metabolism of ductal carcinoma in situ in animal models. CCR2 mediates metabolism and progression of MIND lesions through MET.
With over 60,000 cases diagnosed annually in the US, ductal carcinoma in situ (DCIS) is the most prevalent form of early-stage breast cancer. Because many DCIS cases never progress to invasive ductal carcinomas (IDC), overtreatment remains a significant problem. Up to 20% patients experience disease recurrence, indicating that standard treatments do not effectively treat DCIS for a subset of patients. By understanding the mechanisms of DCIS progression, we can develop new treatment strategies better tailored to patients. The chemokine CCL2 and its receptor CCR2 are known to regulate macrophage recruitment during inflammation and cancer progression. Recent studies indicate that increased CCL2/CCR2 signaling in breast epithelial cells enhance formation of IDC. Here, we characterized the molecular mechanisms important for CCL2/CCR2-mediated DCIS progression. Phospho-protein array profiling revealed that CCL2 stimulated phosphorylation of MET receptor tyrosine kinases in breast cancer cells. Co-immunoprecipitation and proximity ligation assays demonstrated that CCL2-induced MET activity depended on interactions with CCR2 and SRC. Extracellular flux analysis and biochemical assays revealed that CCL2/CCR2 signaling in breast cancer cells enhanced glycolytic enzyme expression and activity. CRISPR knockout and pharmacologic inhibition of MET revealed that CCL2/CCR2-induced breast cancer cell proliferation, survival, migration and glycolysis through MET-dependent mechanisms. In animals, MET inhibitors blocked CCR2-mediated DCIS progression and metabolism. CCR2 and MET were significantly co-expressed in patient DCIS and IDC tissues. In summary, MET receptor activity is an important mechanism for CCL2/CCR2-mediated progression and metabolism of early-stage breast cancer, with important clinical implications.
Collapse
|
11
|
Xie Z, Xia T, Wu D, Che L, Zhang W, Cai X, Liu S. Identification of the key genes in chronic obstructive pulmonary disease by weighted gene co-expression network analysis. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:665. [PMID: 35845513 PMCID: PMC9279780 DOI: 10.21037/atm-22-2523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/08/2022] [Indexed: 11/30/2022]
Abstract
Background Chronic obstructive pulmonary disease (COPD) is prevalent mainly in older adults, especially those who are smokers. It appears to be regulated by multiple genes, but there is some degree of familial clustering. The evidence to date suggests that COPD-associated biomarkers are largely inadequate for disease diagnosis, so we conducted a comprehensive search for more specific genetic markers. Methods We used 3 datasets from the Gene Expression Omnibus (GEO) database. By investigating the biological information [i.e., Gene Ontology, Kyoto Encyclopedia of Genes and Genomes and weighted gene co-expression network analysis (WGCNA)], we filtered out 8 differentially expressed genes (DEGs) and validated the transcript levels of those hub genes in 16HBE cell lines, THP-1 cell lines and lung tissue of COPD patients. Results The 8 hub genes comprised amyloid precursor protein (APP), fibronectin 1, insulin-like growth factor 1 (IGF1), β-actin, capping actin protein of muscle Z-line subunit alpha 2, secreted phosphoprotein 1 (SPP1), catalase (CAT), and colony stimulating factor 2 (CSF2) were selected from among the DEGs. Cigarette smoke extract-stimulated 16HBE cells were found to highly express SPP1, CSF2, and IGF1. In addition, IGF1 levels were increased and IGF1 and APP levels were decreased in CSE-stimulated THP-1 cells. SPP1 and FN1 showed increased expression levels in lung tissue of COPD patients, but the opposite held for APP and CAT. Conclusions We identified 8 hub genes of COPD based on GO, KEGG and WGCNA, which have provided insights into the pathophysiological mechanisms of COPD.
Collapse
Affiliation(s)
- Zhefan Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tingting Xia
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dongxue Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Li Che
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wei Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xingdong Cai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shengming Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
12
|
Garcia-Robledo JE, Rosell R, Ruíz-Patiño A, Sotelo C, Arrieta O, Zatarain-Barrón L, Ordoñez C, Jaller E, Rojas L, Russo A, de Miguel-Pérez D, Rolfo C, Cardona AF. KRAS and MET in non-small-cell lung cancer: two of the new kids on the 'drivers' block. Ther Adv Respir Dis 2022; 16:17534666211066064. [PMID: 35098800 PMCID: PMC8808025 DOI: 10.1177/17534666211066064] [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: 07/21/2021] [Accepted: 10/14/2021] [Indexed: 12/30/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a heterogeneous disease, and therapeutic management has advanced to identify various critical oncogenic mutations that promote lung cancer tumorigenesis. Subsequent studies have developed targeted therapies against these oncogenes in the hope of personalized treatment based on the tumor's molecular genomics. This review presents a comprehensive review of the biology, new therapeutic interventions, and resistance patterns of two well-defined subgroups, tumors with KRAS and MET alterations. We also discuss the status of molecular testing practices for these two key oncogenic drivers, considering the progressive introduction of next-generation sequencing (NGS) and RNA sequencing in regular clinical practice.
Collapse
Affiliation(s)
| | - Rafael Rosell
- Cancer Biology and Precision Medicine Program, Germans Trias i Pujol Research Institute (IGTP)/Dr. Rosell Oncology Institute (IOR), Quirón-Dexeus University Institute, Barcelona, Spain
| | - Alejandro Ruíz-Patiño
- Direction of Research and Education, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC), Bogotá, Colombia
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogotá, Colombia Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogotá, Colombia Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Carolina Sotelo
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogotá, Colombia Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Oscar Arrieta
- Thoracic Oncology Unit and Personalized Oncology Laboratory, National Cancer Institute (INCan), México City, México
| | - Lucia Zatarain-Barrón
- Thoracic Oncology Unit and Personalized Oncology Laboratory, National Cancer Institute (INCan), México City, México
| | - Camila Ordoñez
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogotá, Colombia Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Elvira Jaller
- Department of Internal Medicine, Universidad El Bosque, Bogotá, Colombia
| | - Leonardo Rojas
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogotá, Colombia Department of Clinical Oncology, Clínica Colsanitas, Bogotá, Colombia Clinical and Translational Oncology Group, Clínica del Country, Bogotá, Colombia
| | - Alessandro Russo
- Medical Oncology Unit, A.O. Papardo, Messina, Italy Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Diego de Miguel-Pérez
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christian Rolfo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
13
|
Stanislovas J, Kermorgant S. c-Met-integrin cooperation: Mechanisms, tumorigenic effects, and therapeutic relevance. Front Cell Dev Biol 2022; 10:994528. [PMID: 36330337 PMCID: PMC9624249 DOI: 10.3389/fcell.2022.994528] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
c-Met is a receptor tyrosine kinase which upon activation by its ligand, the hepatocyte growth factor, mediates many important signalling pathways that regulate cellular functions such as survival, proliferation, and migration. Its oncogenic and tumorigenic signalling mechanisms, greatly contributing to cancer development and progression, are well documented. Integrins, heterogeneous adhesion receptors which facilitate cell-extracellular matrix interactions, are important in biomechanically sensitive cell adhesion and motility but also modulate diverse cell behaviour. Here we review the studies which reported cooperation between c-Met and several integrins, particularly β1 and β4, in various cell models including many tumour cell types. From the various experimental models and results analysed, we propose that c-Met-integrin cooperation occurs via inside-out or outside-in signalling. Thus, either c-Met activation triggers integrin activation and cell adhesion or integrin adhesion to its extracellular ligand triggers c-Met activation. These two modes of cooperation require the adhesive function of integrins and mostly lead to cell migration and invasion. In a third, less conventional, mode of cooperation, the integrin plays the role of a signalling adaptor for c-Met, independently from its adhesive property, leading to anchorage independent survival. Recent studies have revealed the influence of endocytic trafficking in c-Met-integrin cooperation including the adaptor function of integrin occurring on endomembranes, triggering an inside-in signalling, believed to promote survival of metastatic cells. We present the evidence of the cooperation in vivo and in human tissues and highlight its therapeutic relevance. A better understanding of the mechanisms regulating c-Met-integrin cooperation in cancer progression could lead to the design of new therapies targeting this cooperation, providing more effective therapeutic approaches than c-Met or integrin inhibitors as monotherapies used in the clinic.
Collapse
Affiliation(s)
- Justas Stanislovas
- Spatial Signalling Group, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Stéphanie Kermorgant
- Spatial Signalling Group, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
14
|
Yang X, Liao HY, Zhang HH. Roles of MET in human cancer. Clin Chim Acta 2021; 525:69-83. [PMID: 34951962 DOI: 10.1016/j.cca.2021.12.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/18/2023]
Abstract
The MET proto-oncogene was first identified in osteosarcoma cells exposed to carcinogens. Although expressed in many normal cells, MET is overexpressed in many human cancers. MET is involved in the initiation and development of various human cancers and mediates proliferation, migration and invasion. Accordingly, MET has been successfully used as a biomarker for diagnosis and prognosis, survival, post-operative recurrence, risk assessment and pathologic grading, as well as a therapeutic target. In addition, recent work indicates that inhibition of MET expression and function has potential clinical benefit. This review summarizes the role, mechanism, and clinical significance of MET in the formation and development of human cancer.
Collapse
Affiliation(s)
- Xin Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Yang Liao
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China.
| |
Collapse
|
15
|
Liu QQ, Zeng XL, Guan YL, Lu JX, Tu K, Liu FY. Verticillin A inhibits colon cancer cell migration and invasion by targeting c-Met. J Zhejiang Univ Sci B 2021; 21:779-795. [PMID: 33043644 DOI: 10.1631/jzus.b2000190] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Verticillin A is a diketopiperazine compound which was previously isolated from Amanita flavorubescens Alk (containing parasitic fungi Hypomyces hyalines (Schw.) Tul.). Here, we initially found, by wound healing assay and Transwell assay in vitro, that verticillin A possesses an inhibitory effect against the migration and invasion of the human colon cancer cell. Subsequently, c-mesenchymal-epithelial transition factor (c-Met) was identified as a molecular target of verticillin A by screening key genes related to cell migration. Verticillin A-mediated c-Met suppression is at the transcriptional level. Further study demonstrated that verticillin A suppressed c-MET phosphorylation and decreased c-MET protein level. In addition, verticillin A inhibited the phosphorylation of c-MET downstream molecules including rat sarcoma (Ras)-associated factor (Raf), extracellular signal-regulated kinase (ERK), and protein kinase B (AKT). Overexpression of Erk partially reversed the verticillin A-mediated anti-metastasis action in the human colon cancer cell. More importantly, verticillin A also inhibited cancer cell metastasis in vivo. Thus, verticillin A can significantly inhibit the migration and invasion of colon cancer cells by targeting c-Met and inhibiting Ras/Raf/mitogen-activated extracellular signal-regulated kinase (MEK)/ERK signaling pathways. Therefore, we determined that verticillin A is a natural compound that can be further developed as an anti-metastatic drug in human cancers.
Collapse
Affiliation(s)
- Qian-Qian Liu
- Deparement of Internal Medicine, Zhejiang University Hospital, Hangzhou 310027, China
| | - Xue-Li Zeng
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yue-Lin Guan
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jing-Xin Lu
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kai Tu
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fei-Yan Liu
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
16
|
Geletu M, Adan H, Niit M, Arulanandam R, Carefoot E, Hoskin V, Sina D, Elliott B, Gunning P, Raptis L. Modulation of Akt vs Stat3 activity by the focal adhesion kinase in non-neoplastic mouse fibroblasts. Exp Cell Res 2021; 411:112731. [PMID: 34270980 DOI: 10.1016/j.yexcr.2021.112731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Adhesion of cells to each other and to the extracellular matrix (ECM) are both required for cellular functions. Cell-to-cell adhesion is mediated by cadherins, and their engagement triggers the activation of Stat3, which offers a potent survival signal. Adhesion to the ECM on the other hand, activates FAK which attracts and activates Src, as well as receptor tyrosine kinases (RTKs), the PI3k/Akt and Ras/Erk pathways. However, the effect of cell density upon FAK and Akt activity has not been examined. We now demonstrate that, interestingly, despite being potent Stat3 activators, Src and RTKs are unable to activate Stat3 in sparsely growing (i.e., without cadherin engagement), non-neoplastic cells attached to the ECM. In contrast, cell aggregation (i.e., cadherin engagement in the absence of adhesion to a solid substratum) was found to activate both Stat3 and Akt. Pharmacologic or genetic reduction of FAK activity abolished Akt activity at low densities, indicating that FAK is an important activator of Akt in this setting. Notably, FAK knockout increased cellular sensitivity to the Stat3 inhibitor CPA7, while FAK reintroduction restored resistance to this drug. These findings suggest a complementary role of integrin/FAK/Akt and cadherin/Stat3-mediated pro-survival pathways, which may be of significance during neoplastic transformation and metastasis.
Collapse
Affiliation(s)
- Mulu Geletu
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Mdicine, Queen's University, Kingston, ON, K7L 3N6, Canada.
| | - Hanad Adan
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Mdicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Maximillian Niit
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Mdicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Rozanne Arulanandam
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Mdicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Esther Carefoot
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Mdicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Victoria Hoskin
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Mdicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Diana Sina
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Mdicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Bruce Elliott
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Mdicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Patrick Gunning
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Mdicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Leda Raptis
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Mdicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| |
Collapse
|
17
|
Yang Y, Lin Y, Zhang Z, Xu R, Yu X, Deng F. Micro/nano-net guides M2-pattern macrophage cytoskeleton distribution via Src-ROCK signalling for enhanced angiogenesis. Biomater Sci 2021; 9:3334-3347. [PMID: 33725044 DOI: 10.1039/d1bm00116g] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Implant surface topography has been proven to determine the fate of adhered macrophage polarization, which is closely related to the cytoskeletal arrangement during adhesion. Our purpose was to establish a topography that is favourable to M2 macrophage switching by regulating macrophage cytoskeleton distribution. Two micro/nano-net structures with different pore sizes were generated by alkali bathing at medium (SAM) or high (SAH) temperature based on the micro-level surface. Their surface characteristics, in vitro macrophage polarization and impact on endothelial cells were analysed. The in vivo macrophage response and osseointegration were also tested. The results showed that the micro/nano-net has high hydrophilicity and moderate roughness. In the SAH and SAM groups, macrophages exhibited an elongated cytoskeleton with tiny protrusions and had a high M2/M1 polarization ratio with enhanced angiogenic ability, and in vivo studies also showed faster angiogenesis and bone formation in these groups. SAH showed even better results than SAM. For cytoskeleton related pathway explanation, ROCK expression was upregulated and Src expression was downregulated at the early or late adhesion stage in both the SAH and SAM groups. These results indicated that the micro/nano-net structure guides elongated macrophage adhesion states via Src-ROCK signalling and switches macrophages towards the M2 phenotype, which provides a cytoskeleton-oriented topography design for an ideal immune response.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Yujing Lin
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Zhengchuan Zhang
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Ruogu Xu
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Xiaoran Yu
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Feilong Deng
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| |
Collapse
|
18
|
Geletu M, Adan H, Niit M, Arulanandam R, Carefoot E, Hoskin V, Sina D, Elliott B, Gunning P, Raptis L. Modulation of Akt vs Stat3 activity by the focal adhesion kinase in non-neoplastic mouse fibroblasts. Exp Cell Res 2021; 404:112601. [PMID: 33957118 DOI: 10.1016/j.yexcr.2021.112601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 12/28/2022]
Abstract
Adhesion of cells to each other and to the extracellular matrix (ECM) are both required for cellular functions. Cell-to-cell adhesion is mediated by cadherins and their engagement triggers the activation of Stat3, which offers a potent survival signal. Adhesion to the ECM on the other hand, activates FAK which attracts and activates Src, as well as receptor tyrosine kinases (RTKs), the PI3k/Akt and Ras/Erk pathways. However, the effect of cell density upon FAK and Akt activity has not been examined. We now demonstrate that, interestingly, despite being potent Stat3 activators, Src and RTKs are unable to activate Stat3 in sparsely growing (i.e., without cadherin engagement), non-neoplastic cells attached to the ECM. In contrast, cell aggregation (i.e., cadherin engagement in the absence of adhesion to a solid substratum) was found to activate both Stat3 and Akt. Pharmacologic or genetic reduction of FAK activity abolished Akt activity at low densities, indicating that FAK is an important activator of Akt in this setting. Notably, FAK knockout increased cellular sensitivity to the Stat3 inhibitor CPA7, while FAK reintroduction restored resistance to this drug. These findings suggest a complementary role of integrin/FAK/Akt and cadherin/Stat3-mediated pro-survival pathways, which may be of significance during neoplastic transformation and metastasis.
Collapse
Affiliation(s)
- Mulu Geletu
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, K7L 3N6, Canada; Department of Chemical and Physical Sciences, University of Toronto, Mississauga, Ontario, L5L 1C6, Canada.
| | - Hanad Adan
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Maximillian Niit
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Rozanne Arulanandam
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, K7L 3N6, Canada; Department of Pathology and Molecular Medicine, Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, K1H 8L6, Canada
| | - Esther Carefoot
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Victoria Hoskin
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Diana Sina
- Department of Chemical and Physical Sciences (CPS), University of Toronto Mississauga, 3359 Mississauga Rd, Mississauga, ON, L5L 1C6, Canada
| | - Bruce Elliott
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Patrick Gunning
- Department of Chemical and Physical Sciences, University of Toronto, Mississauga, Ontario, L5L 1C6, Canada
| | - Leda Raptis
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| |
Collapse
|
19
|
Ólafsson EB, Ten Hoeve AL, Li-Wang X, Westermark L, Varas-Godoy M, Barragan A. Convergent Met and voltage-gated Ca 2+ channel signaling drives hypermigration of Toxoplasma-infected dendritic cells. J Cell Sci 2020; 134:jcs241752. [PMID: 32161101 DOI: 10.1242/jcs.241752] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/26/2020] [Indexed: 01/11/2023] Open
Abstract
Ras-Erk MAPK signaling controls many of the principal pathways involved in metazoan cell motility, drives metastasis of multiple cancer types and is targeted in chemotherapy. However, its putative roles in immune cell functions or in infections have remained elusive. Here, using primary dendritic cells (DCs) in an infection model with the protozoan Toxoplasma gondii, we show that two pathways activated by infection converge on Ras-Erk MAPK signaling to promote migration of parasitized DCs. We report that signaling through the receptor tyrosine kinase Met (also known as HGF receptor) contributes to T. gondii-induced DC hypermotility. Furthermore, voltage-gated Ca2+ channel (VGCC, subtype CaV1.3) signaling impacted the migratory activation of DCs via calmodulin-calmodulin kinase II. We show that convergent VGCC signaling and Met signaling activate the GTPase Ras to drive Erk1 and Erk2 (also known as MAPK3 and MAPK1, respectively) phosphorylation and hypermotility of T. gondii-infected DCs. The data provide a molecular basis for the hypermigratory mesenchymal-to-amoeboid transition (MAT) of parasitized DCs. This emerging concept suggests that parasitized DCs acquire metastasis-like migratory properties that promote infection-related dissemination.
Collapse
Affiliation(s)
- Einar B Ólafsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Arne L Ten Hoeve
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Xiaoze Li-Wang
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Linda Westermark
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Manuel Varas-Godoy
- Cancer Cell Biology Laboratory, Center for Cell Biology and Biomedicine (CEBICEM), Faculty of Medicine and Science, Universidad San Sebastian, 7620001 Santiago, Chile
| | - Antonio Barragan
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| |
Collapse
|
20
|
MET targeting: time for a rematch. Oncogene 2020; 39:2845-2862. [PMID: 32034310 DOI: 10.1038/s41388-020-1193-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/16/2020] [Accepted: 01/24/2020] [Indexed: 12/21/2022]
Abstract
MET, the receptor tyrosine kinase (RTK) for hepatocyte growth factor, is a proto-oncogene involved in embryonic development and throughout life in homeostasis and tissue regeneration. Deregulation of MET signaling has been reported in numerous malignancies, prompting great interest in MET targeting for cancer therapy. The present review offers a summary of the biology of MET and its known functions in normal physiology and carcinogenesis, followed by an overview of the most relevant MET-targeting strategies and corresponding clinical trials, highlighting both past setbacks and promising future prospects. By placing their efforts on a more precise stratification strategy through the genetic analysis of tumors, modern trials such as the NCI-MATCH trial could revive the past enthusiasm for MET-targeted therapy.
Collapse
|
21
|
Xie R, Liu Z, Lin Z, Shi P, Chen B, Li S, Li G, Huang L, Lin X, Yao H. Potential mechanism of action of Ixeris sonchifolia extract injection against cardiovascular diseases revealed by combination of HPLC-Q-TOF-MS, virtual screening and systems pharmacology approach. RSC Adv 2020; 10:38497-38504. [PMID: 35517561 PMCID: PMC9057262 DOI: 10.1039/d0ra07038f] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 10/04/2020] [Indexed: 12/12/2022] Open
Abstract
Ixeris sonchifolia extract injection, a Chinese medicine preparation named as Kudiezi injection (KDZI) in China, has been widely used for the treatment of cardiovascular diseases (CVDs) in recent years. Owing to the component complexity of the preparation, the study on the effect mechanism of the herbal medicine against CVDs is a big challenge. In this research, HPLC-Q-TOF-MS was used to analyze the constituents of the preparation, disclosing that the KDZI mainly consists of 10 ingredients, namely 3-caffeoylquinic acid (KDZI-1), 4-caffeoylquinic acid (KDZI-2), 5-caffeoylquinic acid (KDZI-3), apigenin-7-O-β-d-glucuronide (KDZI-4), caffeic acid (KDZI-5), chicoric acid (KDZI-6), caftaric acid (KDZI-7), luteolin-7-O-β-d-gentiobioside (KDZI-8), luteolin-7-O-β-d-glucopyranoside (KDZI-9) and luteolin-7-O-β-d-glucuronide (KDZI-10). Afterwards, target fishing and an integrated systems pharmacology approach combined with molecular docking (Sybyl 1.3 and AutoDock Vina) were adopted to predict the potential targets and pathways for the main ingredients in KDZI. As results, 39 protein targets and 9 KEGG pathways, possessing high relevance to the therapeutic effects of the ingredients of KDZI against CVDs, were screened out reasonably. The integrated pharmacology analysis suggested that KDZI could exert its therapeutic effects against CVDs possibly via multi-targets including EGFR, MAPK10, and SRC and multi-pathways referring to MAPK, focal adhesion, complement and coagulation cascades, etc. This research provides insights into understanding the comprehensive therapeutic effect and mechanism of the KDZI on CVDs. Ixeris sonchifolia extract injection, a Chinese medicine preparation named as Kudiezi injection (KDZI) in China, has been widely used for the treatment of cardiovascular diseases (CVDs) in recent years.![]()
Collapse
|
22
|
Targeting the COX2/MET/TOPK signaling axis induces apoptosis in gefitinib-resistant NSCLC cells. Cell Death Dis 2019; 10:777. [PMID: 31611604 PMCID: PMC6791885 DOI: 10.1038/s41419-019-2020-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/20/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023]
Abstract
MET overactivation is one of the crucial reasons for tyrosine kinase inhibitor (TKI) resistance, but the mechanisms are not wholly clear. Here, COX2, TOPK, and MET expression were examined in EGFR-activating mutated NSCLC by immunohistochemical (IHC) analysis. The relationship between COX2, TOPK, and MET was explored in vitro and ex vivo. In addition, the inhibition of HCC827GR cell growth by combining COX2 inhibitor (celecoxib), TOPK inhibitor (pantoprazole), and gefitinib was verified ex vivo and in vivo. We found that COX2 and TOPK were highly expressed in EGFR-activating mutated NSCLC and the progression-free survival (PFS) of triple-positive (COX2, MET, and TOPK) patients was shorter than that of triple-negative patients. Then, we observed that the COX2-TXA2 signaling pathway modulated MET through AP-1, resulting in an inhibition of apoptosis in gefitinib-resistant cells. Moreover, we demonstrated that MET could phosphorylate TOPK at Tyr74 and then prevent apoptosis in gefitinib-resistant cells. In line with these findings, the combination of celecoxib, pantoprazole, and gefitinib could induce apoptosis in gefitinib-resistant cells and inhibit tumor growth ex vivo and in vivo. Our work reveals a novel COX2/MET/TOPK signaling axis that can prevent apoptosis in gefitinib-resistant cells and suggests that a triple combination of FDA-approved drugs would provide a low-cost and practical strategy to overcome gefitinib resistance.
Collapse
|
23
|
Lu J, Li X, Tu K, Guan Y, Fung KP, Liu F. Verticillin A suppresses HGF-induced migration and invasion via repression of the c-Met/FAK/Src pathway in human gastric and cervical cancer cells. Onco Targets Ther 2019; 12:5823-5833. [PMID: 31440058 PMCID: PMC6668566 DOI: 10.2147/ott.s208683] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/12/2019] [Indexed: 01/06/2023] Open
Abstract
Background and purpose: Verticillin A is a fungal epipolythiodioxopiperazine (ETP) metabolite that was isolated from Amanita flavorubescens Alk infected by Verticillium sp. It was previously proven to possess potent anti-tumor cell growth activity, and we have recently determined that verticillin A is a selective inhibitor of H3K9me3-specific histone methyltransferase. The objective of this study was to find out whether verticillin A is an effective agent for suppression of gastric and cervical tumor progression. Materials and methods: Wound healing and transwell assays was performed to evaluate the effect of verticillin A on hepatocyte growth factor (HGF)-induced AGS and HeLa cells migration and invasion in vitro. Western blot was used to detect signaling proteins verticillin A affected. Results: We determined that verticillin A effectively suppressed hepatocyte growth factor (HGF)-induced AGS and HeLa cells migration and invasion in vitro. At the molecular level, we demonstrated that verticillin A inhibited HGF-induced c-Met phosphorylation and repressed the expression of total c-Met protein in AGS and HeLa cells, resulting from reduced expression of fatty acid synthase. In addition, verticillin A could suppress c-Met downstream FAK/Src signaling pathways by impairing c-Met phosphorylation induced by HGF. Conclusion: Our study demonstrated verticillin A inhibits the migration ability of human gastric cancer (AGS) cells and cervical cancer (HeLa) cells by targeting c-Met and its downstream FAK/Src signaling pathways, and suggested that verticillin A acts as a novel HGF/c-Met inhibitor by reducing expression of this receptor.
Collapse
Affiliation(s)
- Jingxin Lu
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou310058, People’s Republic of China
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xia Li
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou 310022, People’s Republic of China
| | - Kai Tu
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou310058, People’s Republic of China
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yuelin Guan
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou310058, People’s Republic of China
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
| | - Kwok-Pui Fung
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
- School of Biomedical Sciences (SBS), The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
| | - Feiyan Liu
- Research Centre of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou310058, People’s Republic of China
- Joint Centre of Zhejiang University and the Chinese University of Hong Kong on Natural Products and Toxicology Research, Zhejiang University, Hangzhou, People’s Republic of China
| |
Collapse
|
24
|
Jariwala N, Mendoza RG, Garcia D, Lai Z, Subler MA, Windle JJ, Mukhopadhyay ND, Fisher PB, Chen Y, Sarkar D. Posttranscriptional Inhibition of Protein Tyrosine Phosphatase Nonreceptor Type 23 by Staphylococcal Nuclease and Tudor Domain Containing 1: Implications for Hepatocellular Carcinoma. Hepatol Commun 2019; 3:1258-1270. [PMID: 31497746 PMCID: PMC6719750 DOI: 10.1002/hep4.1400] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/16/2019] [Indexed: 01/12/2023] Open
Abstract
Oncoprotein staphylococcal nuclease and tudor domain containing 1 (SND1) regulates gene expression at a posttranscriptional level in multiple cancers, including hepatocellular carcinoma (HCC). Staphylococcal nuclease (SN) domains of SND1 function as a ribonuclease (RNase), and the tudor domain facilitates protein–oligonucleotide interaction. In the present study, we aimed to identify RNA interactome of SND1 to obtain enhanced insights into gene regulation by SND1. RNA interactome was identified by immunoprecipitation (IP) of RNA using anti‐SND1 antibody from human HCC cells followed by RNA immunoprecipitation sequencing (RIP‐Seq). Among RNA species that showed more than 10‐fold enrichment over the control, we focused on the tumor suppressor protein tyrosine phosphatase nonreceptor type 23 (PTPN23) because its regulation by SND1 and its role in HCC are not known. PTPN23 levels were down‐regulated in human HCC cells versus normal hepatocytes and in human HCC tissues versus normal adjacent liver, as revealed by immunohistochemistry. In human HCC cells, knocking down SND1 increased and overexpression of SND1 decreased PTPN23 protein. RNA binding and degradation assays revealed that SND1 binds to and degrades the 3′‐untranslated region (UTR) of PTPN23 messenger RNA (mRNA). Tetracycline‐inducible PTPN23 overexpression in human HCC cells resulted in significant inhibition in proliferation, migration, and invasion and in vivo tumorigenesis. PTPN23 induction caused inhibition in activation of tyrosine‐protein kinase Met (c‐Met), epidermal growth factor receptor (EGFR), Src, and focal adhesion kinase (FAK), suggesting that, as a putative phosphatase, PTPN23 inhibits activation of these oncogenic kinases. Conclusion: PTPN23 is a novel target of SND1, and our findings identify PTPN23 as a unique tumor suppressor for HCC. PTPN23 might function as a homeostatic regulator of multiple kinases, restraining their activation.
Collapse
Affiliation(s)
- Nidhi Jariwala
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA
| | - Rachel G Mendoza
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA
| | - Dawn Garcia
- Greehey Children's Cancer Research Institute University of Texas Health Science Center San Antonio San Antonio TX
| | - Zhao Lai
- Greehey Children's Cancer Research Institute University of Texas Health Science Center San Antonio San Antonio TX
| | - Mark A Subler
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA
| | - Jolene J Windle
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA.,Massey Cancer Center Virginia Commonwealth University Richmond VA
| | | | - Paul B Fisher
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA.,Massey Cancer Center Virginia Commonwealth University Richmond VA.,Virginia Commonwealth University Institute of Molecular Medicine Virginia Commonwealth University Richmond VA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute University of Texas Health Science Center San Antonio San Antonio TX.,Department of Epidemiology and Biostatistics University of Texas Health Science Center San Antonio San Antonio TX
| | - Devanand Sarkar
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA.,Massey Cancer Center Virginia Commonwealth University Richmond VA.,Virginia Commonwealth University Institute of Molecular Medicine Virginia Commonwealth University Richmond VA
| |
Collapse
|
25
|
PIM1 kinase promotes cell proliferation, metastasis and tumor growth of lung adenocarcinoma by potentiating the c-MET signaling pathway. Cancer Lett 2018; 444:116-126. [PMID: 30583073 DOI: 10.1016/j.canlet.2018.12.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/19/2018] [Accepted: 12/07/2018] [Indexed: 12/18/2022]
Abstract
The proto-oncogene PIM1 plays essential roles in proliferation, survival, metastasis and drug resistance in hematopoietic and solid tumors. Although PIM1 has been shown to be associated with lymph node metastasis and poor prognosis in non-small cell lung cancer, its underlying molecular mechanisms in this context are still unclear. Here we show that PIM1 is frequently overexpressed in lung adenocarcinomas, and its expression level is associated with c-MET expression and poor clinical outcome. We further demonstrate that PIM1 may regulate c-MET expression via phosphorylation of eukaryotic translation initiation factor 4B (eIF4B) on S406. Depletion of PIM1 decreased cell proliferation, migration, invasion and colony formation in vitro, as well as reduced tumor growth in vivo. And these effects were partially abrogated by restoring of c-MET expression. Our study implicates a promising therapeutic approach in lung adenocarcinoma patients with PIM1 and c-MET overexpression.
Collapse
|
26
|
Georges S, Calleja LR, Jacques C, Lavaud M, Moukengue B, Lecanda F, Quillard T, Gabriel MT, Cartron PF, Baud'huin M, Lamoureux F, Heymann D, Ory B. Loss of miR-198 and -206 during primary tumor progression enables metastatic dissemination in human osteosarcoma. Oncotarget 2018; 9:35726-35741. [PMID: 30515265 PMCID: PMC6254661 DOI: 10.18632/oncotarget.26284] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 10/23/2018] [Indexed: 01/04/2023] Open
Abstract
The metastatic dissemination is a complex multistep process by which tumor cells from a primary site enter into the systemic circulation to finally spread at distant sites. Even if this mechanism is rare at the tumor level, it remains the major cause of Osteosarcoma-patients’ relapse and mortality. MicroRNAs (miRNAs) have recently been described as novel epigenetics’ genes’ expression regulators actively implicated in cancer progression and dissemination. The understanding of their implication in the metastatic spreading could help clinicians to improve the outcome of osteosarcoma. We established the miRNA’s expression-profile between primary bone-tumors (PTs), circulating tumor cells (CTCs) and lung metastatic (META) samples from in vivo mice xenograft models. Our results show that the expression level of the miR-198 and -206 was decreased in META samples, in which the expression of the metastasis-related receptor C-Met was up-regulated. Those expression variations were validated in osteosarcoma patient biopsies from matching primary tumors and lung metastasis. We validated in vitro the endogenous miRNAs inhibitory effects on both migration and invasion, as well as we confirmed by luciferase assays that the C-Met receptor is one of their bona-fide targets. The anti-metastatic effect of these miRNAs was also validated in vivo, as their direct injections into the tumors reduce the number of lung-metastases and prolongs the overall survival of the treated animals. All together, our results suggest the absence of the miR-198 and -206 as powerful predictive biomarkers of the tumor cell dissemination and the rationale of their potential therapeutic use in the treatment of Osteosarcoma.
Collapse
Affiliation(s)
- Steven Georges
- INSERM, UMR-S 1238, Nantes 44035, France.,PhyOs, Sarcomes Osseux et Remodelage des Tissus Calcifiés, Université de Nantes, Nantes 44035, France
| | - Lidia Rodriguez Calleja
- INSERM, UMR-S 1238, Nantes 44035, France.,PhyOs, Sarcomes Osseux et Remodelage des Tissus Calcifiés, Université de Nantes, Nantes 44035, France
| | - Camille Jacques
- INSERM, UMR-S 1238, Nantes 44035, France.,PhyOs, Sarcomes Osseux et Remodelage des Tissus Calcifiés, Université de Nantes, Nantes 44035, France
| | - Melanie Lavaud
- INSERM, UMR-S 1238, Nantes 44035, France.,PhyOs, Sarcomes Osseux et Remodelage des Tissus Calcifiés, Université de Nantes, Nantes 44035, France
| | - Brice Moukengue
- INSERM, UMR-S 1238, Nantes 44035, France.,PhyOs, Sarcomes Osseux et Remodelage des Tissus Calcifiés, Université de Nantes, Nantes 44035, France
| | - Fernando Lecanda
- Division of Oncology, Adhesion and Metastasis Laboratory, Center for Applied Medic al Research, University of Navarra, Pamplona, Navarra 31008, Spain
| | - Thibaut Quillard
- INSERM, UMR-S 1238, Nantes 44035, France.,PhyOs, Sarcomes Osseux et Remodelage des Tissus Calcifiés, Université de Nantes, Nantes 44035, France
| | - Marta Tellez Gabriel
- INSERM, UMR-S 1238, Nantes 44035, France.,European Associated Laboratory Sarcoma Research Unit, INSERM, University of Sheffield, Sheffield S10 2TN, UK
| | - Pierre-François Cartron
- Equipe Apoptose et Progression Tumorale, Centre de Recherche en Cancérologie et Immunologie Nantes Angers, CRCINA, INSERM, U1232, Université de Nantes, Université d'Angers, Nantes 44035, France.,LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain 44800, France.,European Associated Laboratory Sarcoma Research Unit, INSERM, University of Sheffield, Sheffield S10 2TN, UK.,Cancéropole Grand-Ouest, Réseau Epigénétique (RepiCGO), France
| | - Marc Baud'huin
- INSERM, UMR-S 1238, Nantes 44035, France.,PhyOs, Sarcomes Osseux et Remodelage des Tissus Calcifiés, Université de Nantes, Nantes 44035, France
| | - François Lamoureux
- INSERM, UMR-S 1238, Nantes 44035, France.,PhyOs, Sarcomes Osseux et Remodelage des Tissus Calcifiés, Université de Nantes, Nantes 44035, France
| | - Dominique Heymann
- Equipe Apoptose et Progression Tumorale, Centre de Recherche en Cancérologie et Immunologie Nantes Angers, CRCINA, INSERM, U1232, Université de Nantes, Université d'Angers, Nantes 44035, France.,European Associated Laboratory Sarcoma Research Unit, INSERM, University of Sheffield, Sheffield S10 2TN, UK
| | - Benjamin Ory
- INSERM, UMR-S 1238, Nantes 44035, France.,PhyOs, Sarcomes Osseux et Remodelage des Tissus Calcifiés, Université de Nantes, Nantes 44035, France.,Cancéropole Grand-Ouest, Réseau Epigénétique (RepiCGO), France
| |
Collapse
|
27
|
Souid S, Elsayed HE, Ebrahim HY, Mohyeldin MM, Siddique AB, Karoui H, El Sayed KA, Essafi-Benkhadir K. 13 1 -Oxophorbine protopheophorbide A from Ziziphus lotus as a novel mesenchymal-epithelial transition factor receptor inhibitory lead for the control of breast tumor growth in vitro and in vivo. Mol Carcinog 2018; 57:1507-1524. [PMID: 29978911 DOI: 10.1002/mc.22874] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/27/2018] [Accepted: 07/03/2018] [Indexed: 12/18/2022]
Abstract
The failure of chemotherapy especially in triple negative breast cancer (TNBC) patients has been correlated with the overexpression of the mesenchymal-epithelial transition factor (c-Met) receptor. Thus, the hepatocyte growth factor (HGF)/c-Met signaling axis has gained considerable attention as a valid molecular target for breast cancer therapy. This study reports for the first time the discovery of the 131 -oxophorbines pheophorbide A and protopheophorbide A along with chlorophyllide A from Ziziphus lotus, an edible typical Tunisian plant, as the potent antiproliferative compounds against the human breast cancer cells MDA-MB-231 and MCF-7. Compared to other compounds, protopheophorbide A exerted the highest light-independent antiproliferative effect against the metastatic TNBC MDA-MB-231 cells (IC50 = 6.5 μM). In silico, this compound targeted the kinase domain of multiple c-Met crystal structures. It potently inhibited the kinase domain phosphorylation of wild and mutant c-Met in Z-LYTE kinase assay. Protopheophorbide A inhibited HGF-induced downstream c-Met-dependent cell proliferation, survival, adhesion and migration through RAF/MEK/ERK and PI3K/PTEN/AKT signaling pathways modulation, ROS generation and activation of JNK and p38 pathways. Interestingly, this compound impaired the ability of the MDA-MB-231 cells to adhere at different extracellular matrix proteins by reducing the HGF-induced expression of integrins αv, β3, α2, and β1. Moreover, protopheophorbide A exhibited anti-migratory properties (IC50 = 2.2 μM) through impacting the expression levels of E-cadherin, vimentin, β-catenin, FAK, Brk, Rac, and Src proteins. Importantly, treatment with protopheophorbide A significantly inhibited the MDA-MB-231 tumor growth in vivo. Our results suggest that protopheophorbide A could be a novel c-Met inhibitory lead with promise to control c-Met/HGF-dependent breast malignancies.
Collapse
Affiliation(s)
- Soumaya Souid
- Institut Pasteur de Tunis, LR11IPT04, LR16IPT04 Laboratoire d'Epidémiologie Moléculaire et Pathologie Expérimentale Appliquée Aux Maladies Infectieuses, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia
| | - Heba E Elsayed
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana
| | - Hassan Y Ebrahim
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana
| | - Mohamed M Mohyeldin
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana.,Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Abu Bakar Siddique
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana
| | - Habib Karoui
- Institut Pasteur de Tunis, LR11IPT04, LR16IPT04 Laboratoire d'Epidémiologie Moléculaire et Pathologie Expérimentale Appliquée Aux Maladies Infectieuses, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia
| | - Khalid A El Sayed
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana
| | - Khadija Essafi-Benkhadir
- Institut Pasteur de Tunis, LR11IPT04, LR16IPT04 Laboratoire d'Epidémiologie Moléculaire et Pathologie Expérimentale Appliquée Aux Maladies Infectieuses, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
28
|
Koo MA, Lee MH, Kwon BJ, Seon GM, Kim MS, Kim D, Nam KC, Park JC. Exogenous ROS-induced cell sheet transfer based on hematoporphyrin-polyketone film via a one-step process. Biomaterials 2018; 161:47-56. [DOI: 10.1016/j.biomaterials.2018.01.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/15/2018] [Accepted: 01/19/2018] [Indexed: 12/14/2022]
|
29
|
Thewke DP, Kou J, Fulmer ML, Xie Q. The HGF/MET Signaling and Therapeutics in Cancer. CURRENT HUMAN CELL RESEARCH AND APPLICATIONS 2018. [DOI: 10.1007/978-981-10-7296-3_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
30
|
Ohnishi Y, Yasui H, Nozaki M, Nakajima M. Molecularly-targeted therapy for the oral cancer stem cells. JAPANESE DENTAL SCIENCE REVIEW 2017; 54:88-103. [PMID: 29755619 PMCID: PMC5944082 DOI: 10.1016/j.jdsr.2017.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 10/23/2017] [Accepted: 11/14/2017] [Indexed: 01/12/2023] Open
Abstract
Human cancer tissues are heterogeneous in nature and become differentiated during expansion of cancer stem cells (CSCs). CSCs initiate tumorigenesis, and are involved in tumor recurrence and metastasis. Furthermore, data show that CSCs are highly resistant to anticancer drugs. Cetuximab, a specific anti-epidermal growth factor receptor (EGFR) monoclonal antibody, is used in cancer treatment. Although development of resistance to cetuximab is well recognized, the underlying mechanisms remain unclear. Lapatinib, a dual inhibitor of epidermal growth factor receptor (EGFR)/ErbB2, has antiproliferative effects and is used to treat patients with ErbB2-positive metastatic breast cancer. In this review, cetuximab and lapatinib-resistant oral squamous cell carcinoma (OSCC) cells proliferation and migration signal transduction passway is discussed by introducing our research.
Collapse
Affiliation(s)
- Yuichi Ohnishi
- Second Department of Oral and Maxillofacial Surgery, Osaka Dental University, Hirakata, Osaka 573-1121, Japan.,Department of Cell Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroki Yasui
- Second Department of Oral and Maxillofacial Surgery, Osaka Dental University, Hirakata, Osaka 573-1121, Japan
| | - Masami Nozaki
- Department of Cell Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masahiro Nakajima
- Second Department of Oral and Maxillofacial Surgery, Osaka Dental University, Hirakata, Osaka 573-1121, Japan
| |
Collapse
|
31
|
A Review of Anti-Angiogenic Targets for Monoclonal Antibody Cancer Therapy. Int J Mol Sci 2017; 18:ijms18081786. [PMID: 28817103 PMCID: PMC5578174 DOI: 10.3390/ijms18081786] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 12/13/2022] Open
Abstract
Tumor angiogenesis is a key event that governs tumor progression and metastasis. It is controlled by the complicated and coordinated actions of pro-angiogenic factors and their receptors that become upregulated during tumorigenesis. Over the past several decades, vascular endothelial growth factor (VEGF) signaling has been identified as a central axis in tumor angiogenesis. The remarkable advent of recombinant antibody technology has led to the development of bevacizumab, a humanized antibody that targets VEGF and is a leading clinical therapy to suppress tumor angiogenesis. However, despite the clinical efficacy of bevacizumab, its significant side effects and drug resistance have raised concerns necessitating the identification of novel drug targets and development of novel therapeutics to combat tumor angiogenesis. This review will highlight the role and relevance of VEGF and other potential therapeutic targets and their receptors in angiogenesis. Simultaneously, we will also cover the current status of monoclonal antibodies being developed to target these candidates for cancer therapy.
Collapse
|
32
|
Yasui H, Ohnishi Y, Nakajima M, Nozaki M. Migration of oral squamous cell carcinoma cells are induced by HGF/c-Met signalling via lamellipodia and filopodia formation. Oncol Rep 2017; 37:3674-3680. [PMID: 28440510 DOI: 10.3892/or.2017.5587] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 04/03/2017] [Indexed: 11/06/2022] Open
Abstract
The activation of receptor tyrosine kinases (RTKs) results in cellular effects including cell proliferation, survival, migration and invasion; RTKs also play an important role in tumourigenesis. It has been reported that EGFR signalling controls the migration of oral squamous cell carcinoma (OSCC) SAS and HSC3 cells but not of HSC4 cells, although the proliferation of HSC4 cells is regulated by EGF/EGFR. In the present study, we investigated the roles of EGFR and the c-Met signalling pathway in cell migration via filopodia and lamellipodia formation, which may be prerequisites for migration. To explore the role of c-Met in cell migration, we inhibited c-Met RTK activity using the c-Met inhibitor SU11274 and activated c-Met using hepatocyte growth factor (HGF) in three OSCC cell lines HSC4, SAS and Ca9-22 and investigated migration potency using a wound healing assay. We showed that inhibition of c-Met significantly suppressed, and activation of c-Met significantly promoted, the migration of OSCC cells. Additionally, the migration of SAS and Ca9-22 cells was inhibited by the EGFR inhibitors AG1478 and cetuximab and promoted by EGF treatment. Moreover, migration potency was correlated with lamellipodia formation. Furthermore, western blot analyses demonstrated that SU11274 decreased and HGF increased lamellipodin protein levels as well as phosphorylated c-Met levels. Collectively, we demonstrated that c-Met signalling induced lamellipodia formation by upregulating lamellipodin, thereby promoting the migration of OSCC cells.
Collapse
Affiliation(s)
- Hiroki Yasui
- Department of Cell Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuichi Ohnishi
- Department of Cell Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masahiro Nakajima
- Second Department of Oral and Maxillofacial Surgery, Osaka Dental University, Hirakata, Osaka 573-1121, Japan
| | - Masami Nozaki
- Department of Cell Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
33
|
Wang DD, Chen Y, Chen ZB, Yan FJ, Dai XY, Ying MD, Cao J, Ma J, Luo PH, Han YX, Peng Y, Sun YH, Zhang H, He QJ, Yang B, Zhu H. CT-707, a Novel FAK Inhibitor, Synergizes with Cabozantinib to Suppress Hepatocellular Carcinoma by Blocking Cabozantinib-Induced FAK Activation. Mol Cancer Ther 2016; 15:2916-2925. [PMID: 27638856 DOI: 10.1158/1535-7163.mct-16-0282] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/10/2016] [Accepted: 08/24/2016] [Indexed: 11/16/2022]
Abstract
Hepatocellular carcinoma is among the leading causes of cancer-related deaths worldwide, and the development of new treatment regimens is urgently needed to improve therapeutic approach. In our study, we found that the combination of a Met inhibitor, cabozantinib, and a novel FAK inhibitor, CT-707, exerted synergistic antitumor effects against hepatocellular carcinoma in vitro and in vivo Interestingly, further studies showed that therapeutic concentrations of cabozantinib increased the phosphorylation of FAK, which might attenuate the antitumor activity of cabozantinib. The simultaneous exposure to CT-707 effectively inhibited the activation of FAK that was induced by cabozantinib, which contributes to the synergistic effect of the combination. Furthermore, cabozantinib increased the mRNA and protein levels of integrin α5, which is a canonical upstream of FAK, and the introduction of cilengitide to block integrin function could abrogate FAK activation by cabozantinib, indicating that cabozantinib upregulated the phosphorylation of FAK in an integrin-dependent manner. Similar synergy was also observed on PHA-665752, another selective MET inhibitor, indicating that this observation might be a common characteristic of MET-targeting strategies. Our findings not only favor the development of the novel FAK inhibitor CT-707 as a therapeutic agent against hepatocellular carcinoma but also provide a new strategy of combining MET and FAK inhibitors to potentiate the anticancer activities of these two types of agents for treating hepatocellular carcinoma patients. Mol Cancer Ther; 15(12); 2916-25. ©2016 AACR.
Collapse
Affiliation(s)
- Dan-Dan Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ying Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zi-Bo Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Fang-Jie Yan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiao-Yang Dai
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Mei-Dan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ji Cao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jian Ma
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Pei-Hua Luo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yong-Xin Han
- Department of Medicinal Chemistry, Centaurus BioPharma Co., Ltd, Beijing, China
| | - Yong Peng
- Department of Discovery Biology, Centaurus BioPharma Co., Ltd, Beijing, China
| | - Ying-Hui Sun
- Department of Discovery Biology, Centaurus BioPharma Co., Ltd, Beijing, China
| | - Hui Zhang
- Department of Discovery Biology, Centaurus BioPharma Co., Ltd, Beijing, China
| | - Qiao-Jun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hong Zhu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
34
|
Mukhopadhyay UK, Cass J, Raptis L, Craig AW, Bourdeau V, Varma S, SenGupta S, Elliott BE, Ferbeyre G. STAT5A is regulated by DNA damage via the tumor suppressor p53. Cytokine 2016; 82:70-9. [PMID: 26876578 DOI: 10.1016/j.cyto.2016.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/21/2016] [Accepted: 01/23/2016] [Indexed: 11/18/2022]
Abstract
Here we report that the STAT5A transcription factor is a direct p53 transcriptional target gene. STAT5A is well expressed in p53 wild type cells but not in p53-null cells. Inhibition of p53 reduces STAT5A expression. DNA damaging agents such as doxorubicin also induced STAT5A expression in a p53 dependent manner. Two p53 binding sites were mapped in the STAT5A gene and named PBS1 and PBS2; these sites were sufficient to confer p53 responsiveness in a luciferase reporter gene. Chromatin immunoprecipitation experiments revealed that PBS2 has constitutive p53 bound to it, while p53 binding to PBS1 required DNA damage. In normal human breast lobules, weak p53 staining correlated with regions of intense STAT5A staining. Interestingly, in a cohort of triple negative breast tumor tissues there was little correlation between regions of p53 and STAT5A staining, likely reflecting a high frequency of p53 mutations that stabilize the protein in these tumors. We thus reveal an unexpected connection between cytokine signaling and p53.
Collapse
Affiliation(s)
- Utpal K Mukhopadhyay
- Division of Cancer Biology and Genetics, Queen's University, Cancer Research Institute, Kingston, Ontario K7L 3N6, Canada
| | - Jamaica Cass
- Division of Cancer Biology and Genetics, Queen's University, Cancer Research Institute, Kingston, Ontario K7L 3N6, Canada
| | - Leda Raptis
- Division of Cancer Biology and Genetics, Queen's University, Cancer Research Institute, Kingston, Ontario K7L 3N6, Canada
| | - Andrew W Craig
- Department of Biochemistry, Queens University, Kingston, Ontario, Canada
| | - Véronique Bourdeau
- Université de Montréal, Département de biochimie, Montréal, Québec H3C 3J7, Canada
| | - Sonal Varma
- Division of Cancer Biology and Genetics, Queen's University, Cancer Research Institute, Kingston, Ontario K7L 3N6, Canada
| | - Sandip SenGupta
- Division of Cancer Biology and Genetics, Queen's University, Cancer Research Institute, Kingston, Ontario K7L 3N6, Canada
| | - Bruce E Elliott
- Division of Cancer Biology and Genetics, Queen's University, Cancer Research Institute, Kingston, Ontario K7L 3N6, Canada.
| | - Gerardo Ferbeyre
- Université de Montréal, Département de biochimie, Montréal, Québec H3C 3J7, Canada.
| |
Collapse
|
35
|
Zhang J, Babic A. Regulation of the MET oncogene: molecular mechanisms. Carcinogenesis 2016; 37:345-55. [PMID: 26905592 DOI: 10.1093/carcin/bgw015] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/29/2016] [Indexed: 12/26/2022] Open
Abstract
The MET oncogene is a predictive biomarker and an attractive therapeutic target for various cancers. Its expression is regulated at multiple layers via various mechanisms. It is subject to epigenetic modifications, i.e. DNA methylation and histone acetylation. Hypomethylation and acetylation of the MET gene have been associated with its high expression in some cancers. Multiple transcription factors including Sp1 and Ets-1 govern its transcription. After its transcription, METmRNA is spliced into multiple species in the nucleus before being transported to the cytoplasm where its translation is modulated by at least 30 microRNAs and translation initiation factors, e.g. eIF4E and eIF4B. METmRNA produces a single chain pro-Met protein of 170 kDa which is cleaved into α and β chains. These two chains are bound together through disulfide bonds to form a heterodimer which undergoes either N-linked or O-linked glycosylation in the Golgi apparatus before it is properly localized in the membrane. Upon interactions with its ligand, i.e. hepatocyte growth factor (HGF), the activity of Met kinase is boosted through various phosphorylation mechanisms and the Met signal is relayed to downstream pathways. The phosphorylated Met is then internalized for subsequent degradation or recycle via proteasome, lysosome or endosome pathways. Moreover, the Met expression is subject to autoregulation and activation by other EGFRs and G-protein coupled receptors. Since deregulation of the MET gene leads to cancer and other pathological conditions, a better understanding of the MET regulation is critical for Met-targeted therapeutics.
Collapse
Affiliation(s)
- Jack Zhang
- Research and Development, Ventana Medical Systems, Inc., a Member of the Roche Group, Oro Valley, AZ 85755, USA
| | - Andy Babic
- Research and Development, Ventana Medical Systems, Inc., a Member of the Roche Group, Oro Valley, AZ 85755, USA
| |
Collapse
|
36
|
Petrini I. Biology of MET: a double life between normal tissue repair and tumor progression. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:82. [PMID: 25992381 DOI: 10.3978/j.issn.2305-5839.2015.03.58] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 01/28/2015] [Indexed: 01/30/2023]
Abstract
MNNG HOS transforming gene (MET) is a class IV receptor tyrosine kinase, expressed on the surface of epithelial cells. The interaction with the hepatocyte grow factor (HGF) induces MET dimerization and the activation of multiple intracellular pathways leading to cell proliferation, anti-apoptosis, morphogenic differentiation, motility, invasion, and angiogenesis. Knock out mice have demonstrated that MET is necessary for normal embryogenesis including the formation of striate muscles, liver and trophoblastic structures. The overexpression of MET and HGF are common in solid tumors and contribute to determine their growth. Indeed, MET has been cloned as a transforming gene from a chemically induced human osteosarcoma cell line and therefore is considered a proto-oncogene. Germline MET mutations are characteristic of hereditary papillary kidney cancers and MET amplification is observed in tumors including lung and gastric adenocarcinomas. The inhibition of MET signaling is the target for specific drugs that are raising exciting expectation for medical treatment of cancer.
Collapse
|
37
|
Mesenchymal stem cells from patients with rheumatoid arthritis display impaired function in inhibiting Th17 cells. J Immunol Res 2015; 2015:284215. [PMID: 25918734 PMCID: PMC4397051 DOI: 10.1155/2015/284215] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess multipotent and immunomodulatory properties and are suggested to be involved in the pathogenesis of immune-related diseases. This study explored the function of bone marrow MSCs from rheumatoid arthritis (RA) patients, focusing on immunomodulatory effects. RA MSCs showed decreased proliferative activity and aberrant migration capacity. No significant differences were observed in cytokine profiles between RA and control MSCs. The effects of RA MSCs on proliferation of peripheral blood mononuclear cells (PBMCs) and distribution of specific CD4(+) T cell subtypes (Th17, Treg, and Tfh cells) were investigated. RA MSCs appeared to be indistinguishable from controls in suppressing PBMC proliferation, decreasing the proportion of Tfh cells, and inducing the polarization of Treg cells. However, the capacity to inhibit Th17 cell polarization was impaired in RA MSCs, which was related to the low expression of CCL2 in RA MSCs after coculture with CD4(+) T cells. These findings indicated that RA MSCs display defects in several important biological activities, especially the capacity to inhibit Th17 cell polarization. These functionally impaired MSCs may contribute to the development of RA disease.
Collapse
|
38
|
Van Der Steen N, Pauwels P, Gil-Bazo I, Castañon E, Raez L, Cappuzzo F, Rolfo C. cMET in NSCLC: Can We Cut off the Head of the Hydra? From the Pathway to the Resistance. Cancers (Basel) 2015; 7:556-73. [PMID: 25815459 PMCID: PMC4491670 DOI: 10.3390/cancers7020556] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/19/2015] [Accepted: 03/05/2015] [Indexed: 01/05/2023] Open
Abstract
In the last decade, the tyrosine kinase receptor cMET, together with its ligand hepatocyte growth factor (HGF), has become a target in non-small cell lung cancer (NSCLC). Signalization via cMET stimulates several oncological processes amongst which are cell motility, invasion and metastasis. It also confers resistance against several currently used targeted therapies, e.g., epidermal growth factor receptor (EGFR) inhibitors. In this review, we will discuss the basic structure of cMET and the most important signaling pathways. We will also look into aberrations in the signaling and the effects thereof in cancer growth, with the focus on NSCLC. Finally, we will discuss the role of cMET as resistance mechanism.
Collapse
Affiliation(s)
- Nele Van Der Steen
- Center for Oncological Research Antwerp, University of Antwerp, Universiteitsplein 1, Wilrijk 2610, Belgium; E-Mails: (N.V.D.S.); (P.P.)
| | - Patrick Pauwels
- Center for Oncological Research Antwerp, University of Antwerp, Universiteitsplein 1, Wilrijk 2610, Belgium; E-Mails: (N.V.D.S.); (P.P.)
- Molecular Pathology Unit, Pathology Department, Antwerp University Hospital, Wilrijkstraat 10, Edegem 2650, Belgium
| | - Ignacio Gil-Bazo
- Department of Oncology, Clínica Universidad de Navarra, Pamplona 31008, Spain; E-Mails: (I.G.-B.); (E.C.)
| | - Eduardo Castañon
- Department of Oncology, Clínica Universidad de Navarra, Pamplona 31008, Spain; E-Mails: (I.G.-B.); (E.C.)
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital, Wilrijkstraat 10, Edegem 2650, Belgium
| | - Luis Raez
- Thoracic Oncology Program, Memorial Cancer Institute, Memorial Health Care System, Pembroke Pines, FL 33024, USA; E-Mail:
| | - Federico Cappuzzo
- Thoracic Oncology Program, Memorial Cancer Institute, Memorial Health Care System, Pembroke Pines, FL 33024, USA; E-Mail:
| | - Christian Rolfo
- Center for Oncological Research Antwerp, University of Antwerp, Universiteitsplein 1, Wilrijk 2610, Belgium; E-Mails: (N.V.D.S.); (P.P.)
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital, Wilrijkstraat 10, Edegem 2650, Belgium
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +32-3-821-3646; Fax: +32-3-825-1592
| |
Collapse
|
39
|
Li L, Li X, Han X, Yang T, Fu J, Zhang Y, Gou W. An ovarian cancer model with positive ER: Reversion of ER antagonist resistance by Src blockade. Oncol Rep 2014; 32:943-50. [PMID: 24968896 DOI: 10.3892/or.2014.3284] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/16/2014] [Indexed: 11/06/2022] Open
Abstract
Approximately 60% of ovarian cancers are positive for the estrogen receptor (ER); however, ER-targeted treatment is disappointing due to drug resistance as compared with breast cancer. In estrogen-sensitive cancers, estrogen activates Src to phosphorylate p27 promoting its degradation and increasing cell cycle progression. Since Src is frequently activated in ovarian cancers, we investigated whether combined Src and ER blockade by saracatinib and fulvestrant would circumvent anti-estrogen resistance. In 20 out of 40 enrolled patients with immunohistochemically ER-positive ovarian cancer, phosphorylated Src (p-Src) at the site of 416 tyrosine was expressed with a propensity for metastasis and a poorer disease-free survival (DFS) at 3 years following ER antagonist treatment. The effects of ER and Src blockade on cell cycle were assayed in estrogen receptor α (ERα)-positive ovarian cancer. We observed that Src activity was fairly greater in anti-estrogen-resistant ovarian cancer cells than that in the anti-estrogen-sensitive cell line. Estrogen activated Src via ER-Src binding and ER translocation from cytoplasm to nucleus. Mitogenesis was mediated via ERα, not ERβ. Combined saracatinib and fulvestrant increased p27 and inhibited cell cycle progression. Furthermore, dual therapy induced autophagy and inhibited ovarian cancer xenograft growth more effectively than monotherapy. Saracatinib facilitated the therapeutic effects of fulvestrant by antagonizing the estrogen-mediated Src activation. These are supportive of further preclinical assessment of combined fulvestrant and saracatinib in patients with ovarian cancer.
Collapse
Affiliation(s)
- Long Li
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaojun Li
- Department of Second Thoracosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaobing Han
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ting Yang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Fu
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yunfeng Zhang
- Department of Second Thoracosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenli Gou
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
40
|
Exendin-4 pretreated adipose derived stem cells are resistant to oxidative stress and improve cardiac performance via enhanced adhesion in the infarcted heart. PLoS One 2014; 9:e99756. [PMID: 24915574 PMCID: PMC4051823 DOI: 10.1371/journal.pone.0099756] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 05/18/2014] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS), which were largely generated after myocardial ischemia, severely impaired the adhesion and survival of transplanted stem cells. In this study, we aimed to determine whether Exendin-4 pretreatment could improve the adhesion and therapeutic efficacy of transplanted adipose derived stem cells (ADSCs) in ischemic myocardium. In vitro, H2O2 was used to provide ROS environments, in which ADSCs pretreated with Exendin-4 were incubated. ADSCs without pretreatment were used as control. Then, cell adhesion and viability were analyzed with time. Compared with control ADSCs, Exendin-4 treatment significantly increased the adhesion of ADSCs in ROS environment, while reduced intracellular ROS and cell injury as determined by dihydroethidium (DHE) staining live/Dead staining, lactate dehydrogenase-release assay and MTT assay. Western Blotting demonstrated that ROS significantly decreased the expression of adhesion-related integrins and integrin-related focal adhesion proteins, which were significantly reversed by Exendin-4 pretreatment and followed by decreases in caspase-3, indicating that Exendin-4 may facilitate cell survival through enhanced adhesion. In vivo, myocardial infarction (MI) was induced by the left anterior descending artery ligation in SD rats. Autologous ADSCs with or without Exendin-4 pretreatment were injected into the border area of infarcted hearts, respectively. Multi-techniques were used to assess the beneficial effects after transplantation. Longitudinal bioluminescence imaging and histological staining revealed that Exendin-4 pretreatment enhanced the survival and differentiation of engrafted ADSCs in ischemic myocardium, accompanied with significant benefits in cardiac function, matrix remodeling, and angiogenesis compared with non-pretreated ADSCs 4 weeks post-transplantation. In conclusion, transplantation of Exendin-4 pretreated ADSCs significantly improved cardiac performance and can be an innovative approach in the clinical perspective.
Collapse
|
41
|
Hou M, Cui J, Liu J, Liu F, Jiang R, Liu K, Wang Y, Yin L, Liu W, Yu B. Angiopoietin-like 4 confers resistance to hypoxia/serum deprivation-induced apoptosis through PI3K/Akt and ERK1/2 signaling pathways in mesenchymal stem cells. PLoS One 2014; 9:e85808. [PMID: 24465718 PMCID: PMC3897528 DOI: 10.1371/journal.pone.0085808] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 12/02/2013] [Indexed: 01/22/2023] Open
Abstract
Angiopoietin-like 4 (ANGPTL4) is a potential anti-apoptotic agent for various cells. We examined the protective effect of ANGPTL4 on hypoxia/serum deprivation (SD)-induced apoptosis of MSCs, as well as the possible mechanisms. MSCs were obtained from rat bone marrow and cultured in vitro. Apoptosis was induced by hypoxia/SD for up to 24 hr, and assessed by flow cytometry and TUNEL assay. Expression levels of Akt, ERK1/2, focal adhesion kinase (FAK), Src, Bcl-2, Bax, cytochrome C and cleaved caspase-3 were detected by Western blotting. Integrin β1 mRNA was detected by qRT-PCR. Mitochondrial membrane potential was assayed using a membrane-permeable dye. Hypoxia/SD-induced apoptosis was significantly attenuated by recombinant rat ANGPTL4 in a concentration dependent manner. Moreover, ANGPTL4 decreased the hypoxia/SD-induced caspase-3 cleavage and the cytochrome C release, but increased the Bcl-2/Bax ratio and the mitochondrial membrane potential. Decreased expression of integrin β1, the ANGPTL4 receptor was observed during hypoxia/SD conditions, however, such decrease was reversed by ANGPTL4. In addition, ANGPTL4 induced integrin β1-associated FAK and Src phosphorylation, which was blocked by anti-integrin β1 antibody. ANGPTL4 also reversed the hypoxia/SD-induced decrease of Akt and ERK 1/2 phosphorylation, and the effect of ANGPTL4 was abolished by inhibitors of either integrins, ERK1/2, or phosphatidylinositol 3-kinase (PI3K). Blocking integrinβ1, Akt or ERK largely attenuated anti-apoptotic effect of ANGPTL4. ANGPTL4 protects MSCs from hypoxia/SD-induced apoptosis by interacting with integrins to stimulate FAK complex, leading to downstream ERK1/2 and PI3K/Akt signaling pathways and mimicking the pathway in which MSCs contact with the extracellular matrix.
Collapse
Affiliation(s)
- Meng Hou
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Province Heilongjiang, China ; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Jinjin Cui
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Province Heilongjiang, China ; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Jingjin Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Province Heilongjiang, China ; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Fang Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Province Heilongjiang, China ; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Rui Jiang
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Province Heilongjiang, China ; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Kai Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Province Heilongjiang, China ; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Yongshun Wang
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Province Heilongjiang, China ; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Li Yin
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Province Heilongjiang, China ; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Wenhua Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Province Heilongjiang, China ; Intensive Care Unit (ICU) Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Bo Yu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Province Heilongjiang, China ; Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| |
Collapse
|
42
|
Qin J, Tang J, Jiao L, Ji J, Chen WD, Feng GK, Gao YH, Zhu XF, Deng R. A diterpenoid compound, excisanin A, inhibits the invasive behavior of breast cancer cells by modulating the integrin β1/FAK/PI3K/AKT/β-catenin signaling. Life Sci 2013; 93:655-63. [DOI: 10.1016/j.lfs.2013.09.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 08/27/2013] [Accepted: 09/04/2013] [Indexed: 11/26/2022]
|
43
|
Abstract
c-MET is a receptor tyrosine kinase that, after binding with its ligand, hepatocyte growth factor, activates a wide range of different cellular signaling pathways, including those involved in proliferation, motility, migration and invasion. Although c-MET is important in the control of tissue homeostasis under normal physiological conditions, it has also been found to be aberrantly activated in human cancers via mutation, amplification or protein overexpression. This paper provides an overview of the c-MET signaling pathway, including its role in the development of cancers, and provides a rationale for targeting the pathway as a possible treatment option.
Collapse
|
44
|
Liu Z, Wang H, Wang Y, Lin Q, Yao A, Cao F, Li D, Zhou J, Duan C, Du Z, Wang Y, Wang C. The influence of chitosan hydrogel on stem cell engraftment, survival and homing in the ischemic myocardial microenvironment. Biomaterials 2012; 33:3093-106. [PMID: 22265788 DOI: 10.1016/j.biomaterials.2011.12.044] [Citation(s) in RCA: 203] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 12/26/2011] [Indexed: 12/19/2022]
Abstract
One challenge of cellular cardiomyoplasty for myocardial infarction (MI) is how to improve MI microenvironment to facilitate stem cell engraftment, survival and homing for myocardial repair. The application of injectable hydrogels is an effective strategy. However, it has not been thoroughly investigated on the role of the injectable scaffolds, in improving MI microenvironment, providing space and guidance for cell survival, engraftment and homing. We explored an injectable chitosan hydrogel for stem cell delivery into ischemic heart and investigated the beneficial effects and mechanisms of the hydrogel. In vitro, H(2)O(2)-treatment was used to mimic reactive oxygen species (ROS) microenvironment. The influence of ROS and protection of chitosan components on adipose-derived mesenchymal stem cells (ADSCs) was analyzed too. In vivo, MI was induced by the left anterior descending artery ligation in SD rats. PBS, chitosan hydrogel, ADSC/PBS and ADSC/chitosan hydrogel were injected into the border of infracted hearts respectively. Multi-techniques were used to assess the beneficial effects of chitosan hydrogel after transplantation. We observed that ROS generated by ischemia would impair ADSC adhesion molecules, including integrin-related adhesion molecules integrin αV and β1, focal adhesion-related molecules p-FAK and p-Src, and corresponding ligands of host myocardium ICAM1 and VCAM1. Chitosan hydrogel could rescue these molecules through ROS scavenging and recruit key chemokine for stem cell homing, such as SDF-1. The results suggest that chitosan hydrogel could improve MI microenvironment, enhance stem cell engraftment, survival and homing in ischemic heart through ROS scavenging and chemokine recruitment, contributing to myocardial repair.
Collapse
Affiliation(s)
- Zhiqiang Liu
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences, Tissue Engineering Research Center, Academy of Military Medical Sciences, 27 Taiping Rd, Beijing 100850, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Guessous F, Yang Y, Johnson E, Marcinkiewicz L, Smith M, Zhang Y, Kofman A, Schiff D, Christensen J, Abounader R. Cooperation between c-Met and focal adhesion kinase family members in medulloblastoma and implications for therapy. Mol Cancer Ther 2011; 11:288-97. [PMID: 22188814 DOI: 10.1158/1535-7163.mct-11-0490] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We previously showed the involvement of the tyrosine kinase receptor c-Met in medulloblastoma malignancy. The nonreceptor tyrosine kinases focal adhesion kinase (FAK) and Pyk2 are key players in the progression of different cancers. However, their role in medulloblastoma malignancy is not well understood. In this study, using a protein array approach, we found that c-Met induces FAK and Pyk2 phosphorylation in medulloblastoma cells. We therefore studied the interactions between c-Met and FAK/Pyk2 and their implications for medulloblastoma therapy. We found that c-Met activates FAK and Pyk2 in several medulloblastoma cell lines. We also found that FAK and Pyk2 mediate the malignant effects of c-Met on medulloblastoma cell proliferation, migration, and invasion. On the basis of these findings, we hypothesized that combined c-Met and FAK inhibitions would have additive effects on the inhibition of medulloblastoma malignancy. To test this hypothesis, we assessed the effects on medulloblastoma malignancy parameters of single or combined treatments of medulloblastoma cells with c-Met and FAK small-molecule kinase inhibitors. We found a significant increase in the inhibitory effect of both inhibitors on medulloblastoma cell migration and cell invasion as compared with single inhibitions (P < 0.05). In addition, oral gavage treatment with c-Met inhibitor of mice bearing medulloblastoma xenografts significantly reduced in vivo tumor growth. Therefore, combining c-Met inhibitors with FAK inhibitors constitutes a new potential strategy for medulloblastoma therapy. Altogether, our study describes a role for FAK and Pyk2 in medulloblastoma malignancy, uncovers new interactions between c-Met and FAK/Pyk2, and proposes for the first time combining anti-c-Met and anti-FAK inhibitors as a new strategy for medulloblastoma therapy.
Collapse
Affiliation(s)
- Fadila Guessous
- Department of Microbiology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lu YC, Chen HC. Involvement of lipid rafts in adhesion-induced activation of Met and EGFR. J Biomed Sci 2011; 18:78. [PMID: 22032640 PMCID: PMC3244112 DOI: 10.1186/1423-0127-18-78] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Accepted: 10/27/2011] [Indexed: 02/03/2023] Open
Abstract
Background Cell adhesion has been shown to induce activation of certain growth factor receptors in a ligand-independent manner. However, the mechanism for such activation remains obscure. Methods Human epidermal carcinoma A431 cells were used as a model to examine the mechanism for adhesion-induced activation of hepatocyte growth factor receptor Met and epidermal growth factor receptor (EGFR). The cells were suspended and replated on culture dishes under various conditions. The phosphorylation of Met at Y1234/1235 and EGFR at Y1173 were used as indicators for their activation. The distribution of the receptors and lipid rafts on the plasma membrane were visualized by confocal fluorescent microscopy and total internal reflection microscopy. Results We demonstrate that Met and EGFR are constitutively activated in A431 cells, which confers proliferative and invasive potentials to the cells. The ligand-independent activation of Met and EGFR in A431 cells relies on cell adhesion to a substratum, but is independent of cell spreading, extracellular matrix proteins, and substratum stiffness. This adhesion-induced activation of Met and EGFR cannot be attributed to Src activation, production of reactive oxygen species, and the integrity of the cytoskeleton. In addition, we demonstrate that Met and EGFR are independently activated upon cell adhesion. However, partial depletion of Met and EGFR prevents their activation upon cell adhesion, suggesting that overexpression of the receptors is a prerequisite for their self-activation upon cell adhesion. Although Met and EGFR are largely distributed in 0.04% Triton-insoluble fractions (i.e. raft fraction), their activated forms are detected mainly in 0.04% Triton-soluble fractions (i.e. non-raft fraction). Upon cell adhesion, lipid rafts are accumulated at the cell surface close to the cell-substratum interface, while Met and EGFR are mostly excluded from the membrane enriched by lipid rafts. Conclusions Our results suggest for the first time that cell adhesion to a substratum may induce a polarized distribution of lipid rafts to the cell-substratum interface, which may allow Met and EGFR to be released from lipid rafts, thus leading to their activation in a ligand-independent manner.
Collapse
Affiliation(s)
- Ying-Che Lu
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | | |
Collapse
|
47
|
Huang C, Park CC, Hilsenbeck SG, Ward R, Rimawi MF, Wang YC, Shou J, Bissell MJ, Osborne CK, Schiff R. β1 integrin mediates an alternative survival pathway in breast cancer cells resistant to lapatinib. Breast Cancer Res 2011; 13:R84. [PMID: 21884573 PMCID: PMC3236347 DOI: 10.1186/bcr2936] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Revised: 05/24/2011] [Accepted: 08/31/2011] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION The overexpression of human epidermal growth factor receptor (HER)-2 in 20% of human breast cancers and its association with aggressive growth has led to widespread use of HER2-targeted therapies, such as trastuzumab (T) and lapatinib (L). Despite the success of these drugs, their efficacy is limited in patients whose tumors demonstrate de novo or acquired resistance to treatment. The β1 integrin resides on the membrane of the breast cancer cell, activating several elements of breast tumor progression including proliferation and survival. METHODS We developed a panel of HER2-overexpressing cell lines resistant to L, T, and the potent LT combination through long-term exposure and validated these models in 3D culture. Parental and L/T/LT-resistant cells were subject to HER2 and β1 integrin inhibitors in 3D and monitored for 12 days, followed by quantification of colony number. Parallel experiments were conducted where cells were either stained for Ki-67 and Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) or harvested for protein and analyzed by immunoblot. Results were subjected to statistical testing using analysis of variance and linear contrasts, followed by adjustment with the Sidak method. RESULTS Using multiple cell lines including BT474 and HCC1954, we reveal that in L and LT resistance, where phosphorylation of EGFR/HER1, HER2, and HER3 are strongly inhibited, kinases downstream of β1 integrin--including focal adhesion kinase (FAK) and Src--are up-regulated. Blockade of β1 by the antibody AIIB2 abrogates this up-regulation and functionally achieves significant growth inhibition of L and LT resistant cells in 3D, without dramatically affecting the parental cells. SiRNA against β1 as well as pharmacologic inhibition of FAK achieve the same growth inhibitory effect. In contrast, trastuzumab-resistant cells, which retain high levels of phosphorylated EGFR/HER1, HER2, and HER3, are only modestly growth-inhibited by AIIB2. CONCLUSIONS Our data suggest that HER2 activity, which is suppressed in resistance involving L but not T alone, dictates whether β1 mediates an alternative pathway driving resistance. Our findings justify clinical studies investigating the inhibition of β1 or its downstream signaling moieties as strategies to overcome acquired L and LT resistance.
Collapse
Affiliation(s)
- Catherine Huang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Catherine C Park
- Department of Radiation Oncology, University of California San Francisco, 1600 Divisadero Street, MZ Bldg R, San Francisco, CA 94143, USA
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Robin Ward
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Mothaffar F Rimawi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Yen-chao Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Jiang Shou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Mina J Bissell
- Department of Cancer & DNA Damage Responses, Lawrence Berkeley National Laboratories, One Cyclotron Road, Berkeley, CA 94720, USA
| | - C Kent Osborne
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| |
Collapse
|
48
|
Organ SL, Tong J, Taylor P, St-Germain JR, Navab R, Moran MF, Tsao MS. Quantitative phospho-proteomic profiling of hepatocyte growth factor (HGF)-MET signaling in colorectal cancer. J Proteome Res 2011; 10:3200-11. [PMID: 21609022 DOI: 10.1021/pr200238t] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is the second leading cause of death from cancer. The MET receptor tyrosine kinase and/or its ligand HGF are frequently amplified or overexpressed in CRC. It is known that tyrosine phosphorylated proteins are involved in progression and metastasis of colorectal cancer; however, little is known about the MET phospho-proteome in CRC. High resolution mass spectrometry was used to characterize immunoaffinity-purified, phosphotyrosine (pY)-containing tryptic peptides of the MET-expressing CRC cell model, DLD1. A total of 266 unambiguously identified pY sites spanning 168 proteins were identified. Quantification of mass spectrometry ion currents identified 161 pY sites, including many not previously linked to MET signaling, that were modulated in abundance by HGF stimulation. Overlay of these data with protein-protein interaction data sets suggested that many of the identified HGF-modulated phospho-proteins may be directly or indirectly associated with MET. Analysis of pY sequence motifs indicated a prevalence of Src family kinase consensus sequences, and reciprocal signaling between Src and MET was confirmed by using selective small molecule inhibitors of these kinases. Therefore, using quantitative phospho-proteomics profiling, kinase modulation by ligand and inhibitors, and data integration, an outline of the MET signaling network was generated for the CRC model.
Collapse
Affiliation(s)
- Shawna L Organ
- Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, Toronto, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Mostowy S, Janel S, Forestier C, Roduit C, Kasas S, Pizarro-Cerdá J, Cossart P, Lafont F. A role for septins in the interaction between the Listeria monocytogenes INVASION PROTEIN InlB and the Met receptor. Biophys J 2011; 100:1949-59. [PMID: 21504731 PMCID: PMC3077699 DOI: 10.1016/j.bpj.2011.02.040] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 02/08/2011] [Accepted: 02/22/2011] [Indexed: 10/18/2022] Open
Abstract
Septins are conserved GTPases that form filaments and are required for cell division. During interphase, septin filaments associate with cellular membrane and cytoskeleton networks, yet the functional significance of these associations have, to our knowledge, remained unknown. We recently discovered that different septins, SEPT2 and SEPT11, regulate the InlB-mediated entry of Listeria monocytogenes into host cells. Here we address the role of SEPT2 and SEPT11 in the InlB-Met interactions underlying Listeria invasion to explore how septins modulate surface receptor function. We observed that differences in InlB-mediated Listeria entry correlated with differences in Met surface expression caused by septin depletion. Using atomic force microscopy on living cells, we show that septin depletion significantly reduced the unbinding force of InlB-Met interaction and the viscosity of membrane tethers at locations where the InlB-Met interaction occurs. Strikingly, the same order of difference was observed for cells in which the actin cytoskeleton was disrupted. Consistent with a proposed role of septins in association with the actin cytoskeleton, we show that cell elasticity is decreased upon septin or actin inactivation. Septins are therefore likely to participate in anchorage of the Met receptor to the actin cytoskeleton, and represent a critical determinant in surface receptor function.
Collapse
Affiliation(s)
- Serge Mostowy
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U604, Paris, France
- Institut National de la Recherche Agronomique, USC2020, Paris, France
| | - Sébastien Janel
- Cellular Microbiology of Infectious Pathogens—Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- University Lille Nord-de-France, Lille, France
| | | | - Charles Roduit
- Laboratory of Physics of the Living Matter, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Sandor Kasas
- Laboratory of Physics of the Living Matter, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Javier Pizarro-Cerdá
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U604, Paris, France
- Institut National de la Recherche Agronomique, USC2020, Paris, France
| | - Pascale Cossart
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U604, Paris, France
- Institut National de la Recherche Agronomique, USC2020, Paris, France
| | - Frank Lafont
- Cellular Microbiology of Infectious Pathogens—Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- University Lille Nord-de-France, Lille, France
| |
Collapse
|
50
|
Identification of subtilase cytotoxin (SubAB) receptors whose signaling, in association with SubAB-induced BiP cleavage, is responsible for apoptosis in HeLa cells. Infect Immun 2010; 79:617-27. [PMID: 21098100 DOI: 10.1128/iai.01020-10] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Subtilase cytotoxin (SubAB), which is produced by certain strains of Shiga-toxigenic Escherichia coli (STEC), causes the 78-kDa glucose-regulated protein (GRP78/BiP) cleavage, followed by induction of endoplasmic reticulum (ER) stress, leading to caspase-dependent apoptosis via mitochondrial membrane damage by Bax/Bak activation. The purpose of the present study was to identify SubAB receptors responsible for HeLa cell death. Four proteins, NG2, α2β1 integrin (ITG), L1 cell adhesion molecule (L1CAM), and hepatocyte growth factor receptor (Met), were identified to be SubAB-binding proteins by immunoprecipitation and purification, followed by liquid chromatography-tandem mass spectrometry analysis. SubAB-induced Bax conformational change, Bax/Bak complex formation, caspase activation, and cell death were decreased in β1 ITG, NG2, and L1CAM small interfering RNA-transfected cells, but unexpectedly, BiP cleavage was still observed. Pretreatment of cells with a function-blocking β1 ITG antibody (monoclonal antibody [MAb] P5D2) enhanced SubAB-induced caspase activation; MAb P5D2 alone had no effect on caspase activation. Furthermore, we found that SubAB induced focal adhesion kinase fragmentation, which was mediated by a proteasome-dependent pathway, and caspase activation was suppressed in the presence of proteasome inhibitor. Thus, β1 ITG serves as a SubAB-binding protein and may interact with SubAB-signaling pathways, leading to cell death. Our results raise the possibility that although BiP cleavage is necessary for SubAB-induced apoptotic cell death, signaling pathways associated with functional SubAB receptors may be required for activation of SubAB-dependent apoptotic pathways.
Collapse
|