1
|
Pol M, Gao H, Fox JM, Jia X. TGFβ1 and RGD Cooperatively Regulate SMAD2/3-Mediated Oncogenic Effects in Prostate Cancer Cells in Bio-Orthogonally Constructed Hydrogels. ACS Biomater Sci Eng 2025; 11:3003-3018. [PMID: 40214406 DOI: 10.1021/acsbiomaterials.5c00007] [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] [Indexed: 05/13/2025]
Abstract
To recapitulate prostate cancer metastasis, DU145 cells were cultured in a hyaluronic acid-based, bio-orthogonally constructed, protease-degradable hydrogels. In the presence of a covalently conjugated integrin-binding peptide (GRGDSP), DU145 cells formed tumoroids and exhibited small protrusions. Upon addition of soluble transforming growth factor beta 1 (TGFβ1), cells underwent morphological changes to form extended interconnected cellular networks. Contrarily, in RGD-free hydrogels, cells maintained spherical structures even in the presence of TGFβ1. In RGD-conjugated hydrogels, TGFβ1 induced nuclear localization of SMAD2/3, upregulating a wide range of TGFβ1 target genes and proteins. Prolonged exposure to TGFβ1 led to matrix remodeling and induced epithelial-to-mesenchymal transition in DU145 cells, with loss of epithelial markers and gain of mesenchymal markers. A pharmacological inhibitor of TGFβRI/ALK5, SB-431542, attenuated TGFβ1-induced morphological changes, abrogated nuclear localization of SMAD2/3, and restored the expression of key epithelial markers. Our findings highlight the cooperative role of TGFβ1 signaling and integrin-binding peptide in the acquisition of an aggressive phenotype and the promotion of tumor progression.
Collapse
Affiliation(s)
- Mugdha Pol
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
| | - Hanyuan Gao
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Joseph M Fox
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Xinqiao Jia
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
- Delaware Biotechnology Institute, Newark, Delaware 19713, United States
| |
Collapse
|
2
|
Abdulaziz O, Khan FR, Alharthi NS, Alhuthali HM, Hazazi A, Alzahrani HA, Gharib AF, Alsalmi OA, Hawsawi NM, Alhazmi AY. Computational insights into overcoming resistance mechanisms in targeted therapies for advanced breast cancer: focus on EGFR and HER2 co-inhibition. J Biomol Struct Dyn 2025; 43:4215-4226. [PMID: 38234016 DOI: 10.1080/07391102.2024.2301766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 12/30/2023] [Indexed: 01/19/2024]
Abstract
In the present study, the formation of a heterodimer involving both epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) has been explored as a potential therapeutic mechanism to inhibit the progression of breast cancer. Virtual screening using molecular docking resulted in the three hit compounds (ZINC08382411, ZINC08382438, and ZINC08382292) with minimum binding scores and commonly binding to both receptors. Further, MD simulation analysis of these complexes illustrated the high stability of these compounds with EGFR and HER2. RMSD showed that ZINC08382411 displayed the most stable RMSD of 2 - 3 Å when bound to both receptors, suggesting to have strong compatibility with the active site of the receptor. Hydrogen bond analysis showed that ZINC08382411 forms the maximum number of H-bonds (2 to 3) in both EGFR and HER2 bound complexes, with the highest occupancy of 62% and 79%, respectively. Binding free energy calculation showed that ZINC08382411 possesses maximum affinity towards both the receptors with ΔGbind = -129.628 and -164.063 kJ/mol, respectively. This approach recognizes the significance of EGFR and HER2 in breast cancer development and aims to disrupt their collaborative signaling, which is known to promote the antagonistic behavior of cancer cells. By focusing on this EGFR/HER2 heterodimer, the study offers a promising avenue for identifying a potential candidate (ZINC08382411) that may inhibit breast cancer cell growth and potentially improve patient outcomes. The study's findings may contribute to the ongoing efforts to advance breast cancer treatment strategies.
Collapse
Affiliation(s)
- Osama Abdulaziz
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif Province, Kingdom of Saudi Arabia
| | - Farhan R Khan
- Department of Clinical Laboratory Science,College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Nahed S Alharthi
- Department of Medical Laboratory, College of Applied Medical Sciences in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Hayaa M Alhuthali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Kingdom of Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Hind A Alzahrani
- Department of Basic Sciences, College of Applied of Medical Sciences, Albaha University, Albaha, Saudi Arabia
| | - Amal F Gharib
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Ohud A Alsalmi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Nahed M Hawsawi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Abdulfattah Y Alhazmi
- Pharmaceutical Practices Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
3
|
Gao B, Sabnis R, Kotnis S, Feliciano S, Poling K, Mei T, Feng M, Das JK, Song J, Sun Q. Modular Platform for Efficient Assembly of Multifunctional Antibodies Using Orthogonal Protein-Protein Interactions. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20685-20692. [PMID: 40159649 PMCID: PMC11986891 DOI: 10.1021/acsami.4c21958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025]
Abstract
Multifunctional antibodies, capable of simultaneously engaging multiple targets, are a unique class of antibodies that have sparked growing interest. Current approaches for making multifunctional antibodies, including chemical conjugation or genetic modifications, suffer from low product yield, complex structure design, and complicated manufacturing processes. In this study, we report a modular post-translational platform with highly specific protein-protein interactions for multifunctional antibody assembly and an elastin-like polypeptide (ELP) for easy purification. We generated and purified multifunctional antibodies with over 90% assembled scaffold and overall product purity. Additionally, we assembled antibodies with diverse applications, including detecting cancer, inhibiting cancer cell growth, and directing T cells to cancer cells for enhanced therapeutic efficacy. This platform offers high assembly efficiency, easy purification, and modularity for the redesign of antibody functions.
Collapse
Affiliation(s)
- Baizhen Gao
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Rushant Sabnis
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Siddhi Kotnis
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Sofia Feliciano
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Kyge Poling
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Tracy Mei
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Min Feng
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Jugal Kishore Das
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas 77807, United States
| | - Jianxun Song
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas 77807, United States
| | - Qing Sun
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
- Interdisciplinary
Graduate Program in Genetics and Genomics, Texas A&M University, College
Station, Texas 77843, United States
| |
Collapse
|
4
|
Li X, Zhang L, Zhang Z, Li X, Qian J, Zhou J, Che H, Han Y, Qi X, Yang F, Huang Y, Peng C, Chen Y, Xing Y, Zhang S. Risks of respiratory and circulatory system diseases induced by exposure to PM 2.5 in high humidity and low solar radiation environments: disease types, genes, and functions. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2025; 47:155. [PMID: 40188413 DOI: 10.1007/s10653-025-02465-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 03/18/2025] [Indexed: 04/08/2025]
Abstract
Epidemiological investigation has found that PM2.5 from high humidity and low solar radiation environments (HHLR-PM2.5) induces the highest premature mortality rates from respiratory and circulatory diseases in China. However, the disease types and pathogenic mechanisms of the respiratory and circulatory diseases induced by HHLR-PM2.5 have not been completely revealed. In this study, we explore the risks of commonly existing diseases induced by HHLR-PM2.5 in the respiratory and circulatory systems. For neoplasms, HHLR-PM2.5 significantly induces malignant mesothelioma and arteriovenous hemangioma, the former through the CDKN1A and KIT genes, and the latter through IL6, blood vessel morphogenesis, and transforming growth factor beta binding. Patent ductus arteriosus-persisting type and chronic thromboembolic pulmonary hypertension are the most prominent cardiopulmonary diseases caused by HHLR-PM2.5, with the key molecular target being ACTA2 for the former and CDH5 for the latter. For congenital, hereditary, and neonatal diseases and abnormalities, HHLR-PM2.5 obviously contributes to bronchopulmonary dysplasia and congenital arteriovenous malformation, the former by targeting HMOX1, response to glucocorticoid, and heparin binding, and the latter by targeting IL6, blood vessel morphogenesis, and transforming growth factor beta binding. This study helps to clarify the risks of HHLR-PM2.5 to the respiratory and circulatory systems, supporting and supplementing epidemiology data.
Collapse
Affiliation(s)
- Xiaomeng Li
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
- Institute of Basic Medicine, North Sichuan Medical College, Nanchongn, 637000, Sichua, China
| | - Liru Zhang
- Institute of Basic Medicine, North Sichuan Medical College, Nanchongn, 637000, Sichua, China
| | - Zhengliang Zhang
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
- School of Public Health, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Xuan Li
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
- School of Public Health, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Jingli Qian
- Department of Traditional Chinese Medicine and Rehabilitation, First People's Hospital of Chongqing Liang Jiang New Area, Chongqing, 401121, China
| | - Jiawei Zhou
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Hanxiong Che
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Yan Han
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Xin Qi
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Fumo Yang
- College of Architecture and Environment, Sichuan University, Chengdu, 610065, China
| | - Yu Huang
- Key Laboratory of Aerosol Chemistry and Physics, Institute of Earth Environment, Chinese Academy of Sciences, Xi'an, 710061, China
| | - Chao Peng
- Chongqing Key Laboratory of Urban Atmospheric Environment Observation and Pollution Prevention, Chongqing Research Academy of Ecological Environmental Sciences, Chongqing, 401121, China
| | - Yang Chen
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Yan Xing
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Shumin Zhang
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
- Institute of Basic Medicine, North Sichuan Medical College, Nanchongn, 637000, Sichua, China.
| |
Collapse
|
5
|
Jansen L, Nachtsheim L, Mayer M, Arolt C, Quaas A, Klußmann JP, Wolber P. [Clinical and molecular epidemiology of malignant salivary gland tumors]. Laryngorhinootologie 2025; 104:87-93. [PMID: 39419038 PMCID: PMC11790319 DOI: 10.1055/a-2373-5741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Salivary gland carcinomas are a rare and heterogeneous group of malignant tumors, accounting for 3-6% of all malignant tumors in the head and neck region. The 1-, 3- and 5-year survival rates are 83%, 69% and 63% respectively. Due to new molecular pathological and genetic findings, new entities are constantly being defined as part of the recurring WHO classification of salivary gland carcinomas, so that the incidence rates of the entities are subject to constant change. The only certain risk factor for the development of salivary gland carcinomas is ionizing radiation. In addition, large tumors, cervical lymph node involvement and perineural sheath involvement significantly worsen the prognosis. Today, molecular pathology is coming to the fore, with which potential targets have been identified that can offer prognosis-improving treatment options, particularly in recurrent or distant metastatic stages. Entity-specific tyrosine kinase inhibitors such as axitinib in adenoid cystic carcinoma or larotrectinib in secretory carcinoma and cross-entity therapies such as HER2 inhibition and androgen deprivation can prolong median and progression-free survival with a favorable side effect profile.
Collapse
Affiliation(s)
- Louis Jansen
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Kopf-Hals-Chirurgie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Lisa Nachtsheim
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Kopf-Hals-Chirurgie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Marcel Mayer
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Kopf-Hals-Chirurgie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Christoph Arolt
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Alexander Quaas
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Jens Peter Klußmann
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Kopf-Hals-Chirurgie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Philipp Wolber
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Kopf-Hals-Chirurgie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| |
Collapse
|
6
|
Pasca Fenesan MM, Cosma AA, Melnic E, Cimpean AM, Cozma GV, Negru AG. Heterogeneity of the Alpha-Smooth Muscle Actin Tumor Score in Breast Cancer Cells Significantly Affects Tumor Invasiveness, Recurrence, and Patient Survival. Cureus 2024; 16:e75908. [PMID: 39698197 PMCID: PMC11652793 DOI: 10.7759/cureus.75908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Alpha-smooth muscle actin (αSMA) has been widely investigated in malignancies, primarily concerning its expression in cancer-associated fibroblasts (CAFs) inside the tumor stroma. Microscopic examination indicates that αSMA expression is not confined to the tumor stromal compartment but is also present in a subset of tumor cells, and this expression correlates with an enhanced invasive phenotype of malignant cells from lung, liver, or ovarian malignancies. Information on actin expression in breast cancer (BC) cells is scarce, and its influence on clinicopathological characteristics remains ambiguous due to conflicting findings in the literature. OBJECTIVE To examine the αSMA tumor score in breast cancer cells utilizing digital image analysis (DIA) methodologies and to critically analyze the varying effects of αSMA tumor score values on clinicopathologic parameters, particularly focusing on tumor cell invasiveness, recurrence, and survival. MATERIALS AND METHODS Double immunostaining for CD34 and αSMA was conducted on 53 breast cancer cases that were thoroughly characterized in relation to clinicopathologic data. Double immunostaining for CD34 and αSMA demonstrated different distribution patterns of both markers in normal and breast cancer tissues. DIA data about αSMA tumor cell density, intensity, tumor score, and histological score were correlated with clinicopathological factors. RESULTS We delineated three unique breast cancer subgroups based on αSMA tumor scores: a 9.43% low-expressing subgroup (αSMA_TSlow, score 4), a 35.07% medium-expressing subgroup (αSMA_TSmed, scores 5 and 6), and a 55.5% high-expressing subgroup (αSMA_TShigh, scores 7 and 8). Stromal immature vessels and tertiary lymphoid structures (TLS) exhibited a strong correlation with αSMA_TSlow, whereas recurrence, perineural, and lymphovascular invasion strongly influenced the αSMA_TSmed and αSMA_TShigh subgroups. The αSMA_TSmed subgroup demonstrated the most heterogeneity with the influence of αSMA-expressing breast cancer cells on tumor size, nodal status, perineural and lymphovascular invasion, menopausal status, recurrence, and survival. Most of the cases from the αSMA_TSlow subgroup had Luminal B and Luminal B-HER2 phenotypes, while triple-negative breast cancer (TNBC) represented one-third of all cases in the αSMA_TShigh subgroup. CONCLUSION αSMA-expressing breast cancer cells variably affect malignant growth, invasion, and recurrence, highly contingent upon their density and expression intensity. The current investigation identified an αSMA_TSmed BC subgroup that appears to promote invasiveness, recurrence, and survival in breast cancer. Our data indicate that αSMA BC-expressing cells play a dual role in BC progression, contingent upon their percentage and expression intensity; however, further research is required to elucidate the factors and mechanisms responsible for their accumulation and/or transdifferentiation in malignant breast tissue.
Collapse
Affiliation(s)
- Mihaela-Maria Pasca Fenesan
- Department of Microscopic Morphology/Histology, Doctoral School in Medicine, Department of Clinical Oncology, Victor Babeș University of Medicine and Pharmacy, Timisoara, ROU
| | - Andrei Alexandru Cosma
- Department of Microscopic Morphology/Histology, Doctoral School in Medicine, Department of Clinical Oncology, Victor Babeș University of Medicine and Pharmacy, Timisoara, ROU
| | - Eugen Melnic
- Department of Pathology, Nicolae Testemitanu State University of Medicine and Pharmacy, Chișinău, MDA
| | - Anca Maria Cimpean
- Department of Microscopic Morphology/Histology, Center of Expertise for Rare Vascular Disease in Children, Emergency Hospital for Children Louis, Center of Genomic Medicine, Research Center for Pharmaco-Toxicological Evaluation, Victor Babeș University of Medicine and Pharmacy, Timisoara, ROU
| | - Gabriel Veniamin Cozma
- Discipline of Surgical Semiology I and Thoracic Surgery, Department of Surgery I, Thoracic Surgery Research Center, Victor Babeș University of Medicine and Pharmacy, Timisoara, ROU
| | - Alina Gabriela Negru
- Department of Cardiology, Victor Babeș University of Medicine and Pharmacy, Timisoara, ROU
| |
Collapse
|
7
|
Kubaichuk K, Seitz T, Bergmann U, Glumoff V, Mennerich D, Kietzmann T. Ubiquitin-specific protease 10 determines colorectal cancer outcome by modulating epidermal growth factor signaling via inositol polyphosphate-4-phosphatase type IIB. Oncogenesis 2024; 13:37. [PMID: 39394169 PMCID: PMC11479595 DOI: 10.1038/s41389-024-00538-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024] Open
Abstract
Although there have been advances in understanding colorectal cancer (CRC) pathogenesis, significant gaps still exist, highlighting the need for deeper insights. Dysregulated protein homeostasis, including perturbations in the epidermal growth factor receptor (EGFR) pathway, remains a focal point in CRC pathogenesis. Within this context, the roles of ubiquitin ligases and deubiquitinases have attracted attention, but exploration of their precise contributions is still in its early stages. To address this gap, we investigated the involvement of the deubiquitinase USP10 in CRC. Our in vitro and in vivo study reveals a new paradigm in CRC biology and unravels a novel mechanistic axis, demonstrating for the first time the involvement of inositol polyphosphate 4-phosphatase type II B (INPP4B) in USP10-mediated CRC modulation. Specifically, our study demonstrates that the loss of USP10 results in reduced sensitivity to the EGFR tyrosine kinase inhibitors gefitinib and osimertinib. This is accompanied by a decrease in the activation of the AKT1/PKB pathway upon EGF stimulation, which is mediated by INPP4B. Importantly, in vivo xenograft experiments validate these findings and highlight the crucial role of USP10, particularly in conjunction with INPP4B, in driving CRC progression. The findings enhance our understanding of CRC pathobiology and reveal a new regulatory axis involving USP10 and INPP4B in CRC progression. This unique insight identifies USP10 and INPP4B as potential therapeutic targets in CRC.
Collapse
Affiliation(s)
- Kateryna Kubaichuk
- Faculty of Biochemistry and Molecular Biology, University of Oulu, Oulu, Finland.
| | - Timo Seitz
- Institute of Biomedicine, Faculty of Medicine, Food Sciences Unit, Faculty of Technology, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Ulrich Bergmann
- Faculty of Biochemistry and Molecular Biology, University of Oulu, Oulu, Finland
| | - Virpi Glumoff
- Faculty of Biochemistry and Molecular Biology, University of Oulu, Oulu, Finland
| | - Daniela Mennerich
- Faculty of Biochemistry and Molecular Biology, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Biology, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
8
|
Chen Q, Pan Y, Hu Y, Chen G, Chen X, Xie Y, Wang M, Li Z, Huang J, Shi Y, Huang H, Zhang T, Wang M, Zeng P, Wang S, Chen R, Zheng Y, Zhong L, Yang H, Liang D. An L-type calcium channel blocker nimodipine exerts anti-fibrotic effects by attenuating TGF-β1 induced calcium response in an in vitro model of thyroid eye disease. EYE AND VISION (LONDON, ENGLAND) 2024; 11:37. [PMID: 39237996 PMCID: PMC11378575 DOI: 10.1186/s40662-024-00401-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 08/02/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Thyroid eye disease (TED) is a vision-threatening autoimmune disorder. Orbital tissue fibrosis leading to intractable complications remains a troublesome issue in TED management. Exploration of novel therapeutic targets and agents to ameliorate tissue fibrosis is crucial for TED. Recent work suggests that Ca2+ signaling participates in tissue fibrosis. However, whether an alteration of Ca2+ signaling has a role in fibrogenesis during TED remains unclear. In this study, we aimed to investigate the role of Ca2+ signaling in the fibrogenesis process during TED and the potential therapeutic effects of a highly selective inhibitor of the L-type calcium channel (LTCC), nimodipine, through a TGF-β1 induced in vitro TED model. METHODS Primary culture of orbital fibroblasts (OFs) were established from orbital adipose connective tissues of patients with TED and healthy control donors. Real-time quantitative polymerase chain reaction (RT-qPCR) and RNA sequencing were used to assess the genes expression associated with LTCC in OFs. Flow cytometry, RT-qPCR, 5-ethynyl-2'-deoxyuridine (EdU) proliferation assay, wound healing assay and Western blot (WB) were used to assess the intracellular Ca2+ response on TGF-β1 stimulation, and to evaluate the potential therapeutic effects of nimodipine in the TGF-β1 induced in vitro TED model. The roles of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and signal transducer and activator of transcription 1 (STAT1) in fibrogenesis during TED were determined by immunohistochemistry, WB, flow cytometry and co-immunoprecipitation assay. Selective inhibitors were used to explore the downstream signaling pathways. RESULTS LTCC inhibitor nimodipine blocked the TGF-β1 induced intracellular Ca2+ response and further reduced the expression of alpha-smooth muscle actin (α-SMA), collagen type I alpha 1 (Col1A1) and collagen type I alpha 2 (Col1A2) in OFs. Besides, nimodipine inhibited cell proliferation and migration of OFs. Moreover, our results provided evidence that activation of the CaMKII/STAT1 signaling pathway was involved in fibrogenesis during TED, and nimodipine inhibited the pro-fibrotic functions of OFs by down-regulating the CaMKII/STAT1 signaling pathway. CONCLUSIONS TGF-β1 induces an LTCC-mediated Ca2+ response, followed by activation of CaMKII/STAT1 signaling pathway, which promotes the pro-fibrotic functions of OFs and participates in fibrogenesis during TED. Nimodipine exerts potent anti-fibrotic benefits in vitro by suppressing the CaMKII/STAT1 signaling pathway. Our work deepens our understanding of the fibrogenesis process during TED and provides potential therapeutic targets and alternative candidate for TED.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
- Department of Ophthalmology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Yuan Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Yunwei Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
- Ophthalmic Center, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Guanyu Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Xiaoqing Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Yanyan Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Minzhen Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Zhuang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Jun Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
- Ophthalmic Center, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yuxun Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Haixiang Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Te Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Mei Wang
- Department of Ophthalmology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Peng Zeng
- Department of Ophthalmology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Sha Wang
- Eye Center of Xiangya Hospital, Central South University, Hunan Key Laboratory of Ophthalmology, Changsha, 410008, China
| | - Rongxin Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Yongxin Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Liuxueying Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Huasheng Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Dan Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China.
| |
Collapse
|
9
|
Fu Z, Sun G, Li J, Yu H. Identification of hub genes related to metastasis and prognosis of osteosarcoma and establishment of a prognostic model with bioinformatic methods. Medicine (Baltimore) 2024; 103:e38470. [PMID: 38847690 PMCID: PMC11155596 DOI: 10.1097/md.0000000000038470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 05/15/2024] [Indexed: 06/10/2024] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor occurring in children and adolescents. Improvements in our understanding of the OS pathogenesis and metastatic mechanism on the molecular level might lead to notable advances in the treatment and prognosis of OS. Biomarkers related to OS metastasis and prognosis were analyzed and identified, and a prognostic model was established through the integration of bioinformatics tools and datasets in multiple databases. 2 OS datasets were downloaded from the Gene Expression Omnibus database for data consolidation, standardization, batch effect correction, and identification of differentially expressed genes (DEGs); following that, gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on the DEGs; the STRING database was subsequently used for protein-protein interaction (PPI) network construction and identification of hub genes; hub gene expression was validated, and survival analysis was conducted through the employment of the TARGET database; finally, a prognostic model was established and evaluated subsequent to the screening of survival-related genes. A total of 701 DEGs were identified; by gene ontology and KEGG pathway enrichment analyses, the overlapping DEGs were enriched for 249 biological process terms, 13 cellular component terms, 35 molecular function terms, and 4 KEGG pathways; 13 hub genes were selected from the PPI network; 6 survival-related genes were identified by the survival analysis; the prognostic model suggested that 4 genes were strongly associated with the prognosis of OS. DEGs related to OS metastasis and survival were identified through bioinformatics analysis, and hub genes were further selected to establish an ideal prognostic model for OS patients. On this basis, 4 protective genes including TPM1, TPM2, TPM3, and TPM4 were yielded by the prognostic model.
Collapse
Affiliation(s)
- Zheng Fu
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
- Department of Orthopedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Guofeng Sun
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
| | - Jingtian Li
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
| | - Hongjian Yu
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
| |
Collapse
|
10
|
Long L, Fei X, Chen L, Yao L, Lei X. Potential therapeutic targets of the JAK2/STAT3 signaling pathway in triple-negative breast cancer. Front Oncol 2024; 14:1381251. [PMID: 38699644 PMCID: PMC11063389 DOI: 10.3389/fonc.2024.1381251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Triple-negative breast cancer (TNBC) poses a significant clinical challenge due to its propensity for metastasis and poor prognosis. TNBC evades the body's immune system recognition and attack through various mechanisms, including the Janus Kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. This pathway, characterized by heightened activity in numerous solid tumors, exhibits pronounced activation in specific TNBC subtypes. Consequently, targeting the JAK2/STAT3 signaling pathway emerges as a promising and precise therapeutic strategy for TNBC. The signal transduction cascade of the JAK2/STAT3 pathway predominantly involves receptor tyrosine kinases, the tyrosine kinase JAK2, and the transcription factor STAT3. Ongoing preclinical studies and clinical research are actively investigating this pathway as a potential therapeutic target for TNBC treatment. This article comprehensively reviews preclinical and clinical investigations into TNBC treatment by targeting the JAK2/STAT3 signaling pathway using small molecule compounds. The review explores the role of the JAK2/STAT3 pathway in TNBC therapeutics, evaluating the benefits and limitations of active inhibitors and proteolysis-targeting chimeras in TNBC treatment. The aim is to facilitate the development of novel small-molecule compounds that target TNBC effectively. Ultimately, this work seeks to contribute to enhancing therapeutic efficacy for patients with TNBC.
Collapse
Affiliation(s)
- Lin Long
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiangyu Fei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Liucui Chen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Liang Yao
- Department of Pharmacy, Central Hospital of Hengyang, Hengyang, China
| | - Xiaoyong Lei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
11
|
Hoshimaru T, Nonoguchi N, Kosaka T, Furuse M, Kawabata S, Yagi R, Kurisu Y, Kashiwagi H, Kameda M, Takami T, Kataoka-Sasaki Y, Sasaki M, Honmou O, Hiramatsu R, Wanibuchi M. Actin Alpha 2, Smooth Muscle (ACTA2) Is Involved in the Migratory Potential of Malignant Gliomas, and Its Increased Expression at Recurrence Is a Significant Adverse Prognostic Factor. Brain Sci 2023; 13:1477. [PMID: 37891844 PMCID: PMC10605410 DOI: 10.3390/brainsci13101477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Malignant glioma is a highly invasive tumor, and elucidating the glioma invasion mechanism is essential for developing novel therapies. We aimed to highlight actin alpha 2, smooth muscle (ACTA2) as potential biomarkers of brain invasion and distant recurrence in malignant gliomas. Using the human malignant glioma cell line, U251MG, we generated ACTA2 knockdown (KD) cells treated with small interfering RNA, and the cell motility and proliferation of the ACTA2 KD group were analyzed. Furthermore, tumor samples from 12 glioma patients who underwent reoperation at the time of tumor recurrence were utilized to measure ACTA2 expression in the tumors before and after recurrence. Thereafter, we examined how ACTA2 expression correlates with the time to tumor recurrence and the mode of recurrence. The results showed that the ACTA2 KD group demonstrated a decline in the mean motion distance and proliferative capacity compared to the control group. In the clinical glioma samples, ACTA2 expression was remarkably increased in recurrent samples compared to the primary samples from the same patients, and the higher the change in ACTCA2 expression from the start to relapse, the shorter the progression-free survival. In conclusion, ACTA2 may be involved in distant recurrence in clinical gliomas.
Collapse
Affiliation(s)
- Takumi Hoshimaru
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Naosuke Nonoguchi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Takuya Kosaka
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Motomasa Furuse
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Shinji Kawabata
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Ryokichi Yagi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Yoshitaka Kurisu
- Department of Pathology, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Hideki Kashiwagi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Masahiro Kameda
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Toshihiro Takami
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Yuko Kataoka-Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Hokkaido 060-8556, Japan
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Hokkaido 060-8556, Japan
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Hokkaido 060-8556, Japan
| | - Ryo Hiramatsu
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Masahiko Wanibuchi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| |
Collapse
|
12
|
Li Y, Tan R, Li R, Tian R, Liu Z, Wang X, Chen E, Pan T, Qu H. PKM2/STAT1-mediated PD-L1 upregulation on neutrophils during sepsis promotes neutrophil organ accumulation by serving an anti-apoptotic role. J Inflamm (Lond) 2023; 20:16. [PMID: 37131151 PMCID: PMC10155438 DOI: 10.1186/s12950-023-00341-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/25/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Delayed neutrophil apoptosis during sepsis may impact neutrophil organ accumulation and tissue immune homeostasis. Elucidating the mechanisms underlying neutrophil apoptosis may help identify potential therapeutic targets. Glycolysis is critical to neutrophil activities during sepsis. However, the precise mechanisms through which glycolysis regulates neutrophil physiology remain under-explored, especially those involving the non-metabolic functions of glycolytic enzymes. In the present study, the impact of programmed death ligand-1 (PD-L1) on neutrophil apoptosis was explored. The regulatory effect of the glycolytic enzyme, pyruvate kinase M2 (PKM2), whose role in septic neutrophils remains unaddressed, on neutrophil PD-L1 expression was also explored. METHODS Peripheral blood neutrophils were isolated from patients with sepsis and healthy controls. PD-L1 and PKM2 levels were determined by flow cytometry and Western blotting, respectively. Dimethyl sulfoxide (DMSO)-differentiated HL-60 cells were stimulated with lipopolysaccharide (LPS) as an in vitro simulation of septic neutrophils. Cell apoptosis was assessed by annexin V/propidium iodide (annexin V/PI) staining, as well as determination of protein levels of cleaved caspase-3 and myeloid cell leukemia-1 (Mcl-1) by Western blotting. An in vivo model of sepsis was constructed by intraperitoneal injection of LPS (5 mg/kg) for 16 h. Pulmonary and hepatic neutrophil infiltration was assessed by flow cytometry or immunohistochemistry. RESULTS PD-L1 level was elevated on neutrophils under septic conditions. Administration of neutralizing antibodies against PD-L1 partially reversed the inhibitory effect of LPS on neutrophil apoptosis. Neutrophil infiltration into the lung and liver was also reduced in PD-L1-/- mice 16 h after sepsis induction. PKM2 was upregulated in septic neutrophils and promoted neutrophil PD-L1 expression both in vitro and in vivo. In addition, PKM2 nuclear translocation was increased after LPS stimulation, which promoted PD-L1 expression by directly interacting with and activating signal transducer and activator of transcription 1 (STAT1). Inhibition of PKM2 activity or STAT1 activation also led to increased neutrophil apoptosis. CONCLUSION In this study, a PKM2/STAT1-mediated upregulation of PD-L1 on neutrophils and the anti-apoptotic effect of upregulated PD-L1 on neutrophils during sepsis were identified, which may result in increased pulmonary and hepatic neutrophil accumulation. These findings suggest that PKM2 and PD-L1 could serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Yinjiaozhi Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Ruoming Tan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Ranran Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Rui Tian
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Zhaojun Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Xiaoli Wang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Erzhen Chen
- Department of Emergency Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Tingting Pan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| | - Hongping Qu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| |
Collapse
|
13
|
Park S, Karalis JD, Hong C, Clemenceau JR, Porembka MR, Kim IH, Lee SH, Wang SC, Cheong JH, Hwang TH. ACTA2 Expression Predicts Survival and Is Associated with Response to Immune Checkpoint Inhibitors in Gastric Cancer. Clin Cancer Res 2023; 29:1077-1085. [PMID: 36508166 PMCID: PMC10173146 DOI: 10.1158/1078-0432.ccr-22-1897] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/24/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE We sought to identify biomarkers that predict overall survival (OS) and response to immune checkpoint inhibitors (ICI) for patients with gastric cancer. EXPERIMENTAL DESIGN This was a retrospective study of multiple independent cohorts of patients with gastric cancer. The association between tumor ACTA2 expression and OS and ICI response were determined in patients whose tumors were analyzed with bulk mRNA sequencing. Single-cell RNA sequencing (scRNA-seq) and digital spatial profiling data were used to compare tumors from patients with gastric cancer who did and did not respond to ICI. RESULTS Increasing tumor ACTA2 expression was independently associated with worse OS in a 567-patient discovery cohort [HR, 1.28 per unit increase; 95% confidence interval (CI), 1.02-1.62]. This finding was validated in three independent cohorts (n = 974; HR, 1.52 per unit increase; 95% CI, 1.34-1.73). Of the 108 patients treated with ICI, 56% of patients with low ACTA2 expression responded to ICI versus 25% of patients with high ACTA2 expression (P = 0.004). In an analysis of a publicly available scRNA-seq dataset of 5 microsatellite instability-high patients treated with ICI, the patient who responded to ICI had lower tumor stromal ACTA2 expression than the 4 nonresponders. Digital spatial profiling of tumor samples from 4 ICI responders and 5 ICI nonresponders revealed that responders may have lower ACTA2 expression in α-SMA-positive cancer-associated fibroblasts (CAF) than nonresponders (median: 5.00 vs. 5.50). CONCLUSIONS ACTA2 expression is associated with survival and ICI response in patients with gastric cancer. ACTA2 expression in CAFs, but not in other cellular compartments, appears to be associated with ICI response.
Collapse
Affiliation(s)
- Sunho Park
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL
| | - John D. Karalis
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Changjin Hong
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL
| | - Jean R. Clemenceau
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL
| | - Matthew R. Porembka
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - In-Ho Kim
- Department of Internal Medicine, Division of Medical Oncology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sam C. Wang
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jae-Ho Cheong
- Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, South Korea
| | - Tae Hyun Hwang
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL
- Department of Immunology, Mayo Clinic, Jacksonville, FL
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL
| |
Collapse
|
14
|
Rosado-Galindo H, Domenech M. Surface roughness modulates EGFR signaling and stemness of triple-negative breast cancer cells. Front Cell Dev Biol 2023; 11:1124250. [PMID: 36968199 PMCID: PMC10030610 DOI: 10.3389/fcell.2023.1124250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
Introduction: Cancer stem cells (CSC), a major culprit of drug-resistant phenotypes and tumor relapse, represent less than 2 % of the bulk of TNBC cells, making them difficult to isolate, study, and thus, limiting our understanding of the pathogenesis of the disease. Current methods for CSC enrichment, such as 3D spheroid culture, genetic modification, and stem cell conditioning, are time consuming, expensive, and unsuitable for high-throughput assays. One way to address these limitations is to use topographical stimuli to enhance CSC populations in planar culture. Physical cues in the breast tumor microenvironment can influence cell behavior through changes in the mechanical properties of the extracellular matrix (ECM). In this study, we used topographical cues on polystyrene films to investigate their effect on the proteome and stemness of standard TNBC cell lines. Methods: The topographical polystyrene-based array was generated using razor printing and polishing methods. Proteome data were analyzed and enriched bioprocesses were identified using R software. Stemness was assessed measuring CD44, CD24 and ALDH markers using flow cytometry, immunofluorescence, detection assays, and further validated with mammosphere assay. EGF/EGFR expression and activity was evaluated using enzyme-linked immunosorbent assay (ELISA), immunofluorescence and antibody membrane array. A dose-response assay was performed to further investigate the effect of surface topography on the sensitivity of cells to the EGFR inhibitor. Results: Surface roughness enriched the CSC population and modulated epidermal growth factor receptor (EGFR) signaling activity in TNBC cells. Enhanced proliferation of MDA-MB-468 cells in roughness correlated with upregulation of the epidermal growth factor (EGF) ligand, which in turn corresponded with a 3-fold increase in the expression of EGFR and a 42% increase in its phosphorylation compared to standard smooth culture surfaces. The results also demonstrated that phenotypic changes associated with topographical (roughness) stimuli significantly decreased the drug sensitivity to the EGFR inhibitor gefitinib. In addition, the proportion of CD44+/CD24-/ALDH+ was enhanced on surface roughness in both MDA-MB-231 and MDA-MB-468 cell lines. We also demonstrated that YAP/TAZ activation decreased in a roughness-dependent manner, confirming the mechanosensing effect of the topographies on the oncogenic activity of the cells. Discussion: Overall, this study demonstrates the potential of surface roughness as a culture strategy to influence oncogenic activity in TNBC cells and enrich CSC populations in planar cultures. Such a culture strategy may benefit high-throughput screening studies seeking to identify compounds with broader tumor efficacy.
Collapse
Affiliation(s)
| | - Maribella Domenech
- Bioengineering Program, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
| |
Collapse
|
15
|
Akrida I, Mulita F. The clinical significance of HER2 expression in DCIS. Med Oncol 2022; 40:16. [PMID: 36352293 DOI: 10.1007/s12032-022-01876-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/15/2022] [Indexed: 11/11/2022]
Abstract
HER2 is an established prognostic and predictive marker for patients with invasive breast cancer. The clinical and biological significance of HER2 overexpression in patients with ductal carcinoma in situ (DCIS) remains poorly defined. DCIS is a heterogeneous disease and some patients with DCIS will not progress to invasive breast cancer. However, clinically significant recurrence rates have been reported after breast-conserving surgery for DCIS and approximately half of these cases will be life-threatening invasive recurrences. Since the incidence of DCIS is rising due to the widespread use of screening mammography, there is robust interest in selecting high-risk DCIS patients that may benefit from adjuvant therapies. Molecular prognostic and predictive models in early invasive breast cancer help clinicians identify patients that will benefit from chemotherapy. Molecular subtyping and profiling could also be useful in treating DCIS patients. According to current practice guidelines, HER2 testing is not recommended in DCIS patients. Nevertheless, evidence suggests that HER2-positive DCIS cases may be associated with adverse clinicopathological parameters and increased recurrence rates. This review summarizes the existing body of evidence linking HER2 expression and ipsilateral breast cancer recurrence in DCIS. HER2, as well as its correlation with other clinicopathological markers might be a useful prognostic and predictive marker, helping clinical decision-making in DCIS patients.
Collapse
Affiliation(s)
- Ioanna Akrida
- Department of General Surgery, University General Hospital of Patras, Rion, Greece.
- Department of Anatomy-Histology-Embryology, University of Patras Medical School, 26504, Rion, Greece.
| | - Francesk Mulita
- Department of General Surgery, University General Hospital of Patras, Rion, Greece
| |
Collapse
|
16
|
Abstract
The epidermal growth factor (EGF) system has allowed chemists, biologists, and clinicians to improve our understanding of cell production and cancer therapy. The discovery of EGF led to the recognition of cell surface receptors capable of controlling the proliferation and survival of cells. The detailed structures of the EGF-like ligand and the responses of their receptors (EGFR-family) has revealed the conformational and aggregation changes whereby ligands activate the intracellular kinase domains. Biophysical analysis has revealed the preformed clustering of different EGFR-family members and the processes which occur on ligand binding. Understanding these receptor activation processes and the consequential cytoplasmic signaling has allowed the development of inhibitors which are revolutionizing cancer therapy. This Review describes the recent progress in our understanding of the activation of the EGFR-family, the effects of signaling from the EGFR-family on cell proliferation, and the targeting of the EGFR-family in cancer treatment.
Collapse
Affiliation(s)
- Antony W Burgess
- Honorary Laboratory Head, Personalized Oncology Division, WEHI, Parkville3050, Australia.,Professor Emeritus, Departments of Medical Biology and Surgery (Royal Melbourne Hospital), University of Melbourne, Melbourne3052, Australia.,The Brain Cancer Centre at WEHI, Parkville3052, Australia
| |
Collapse
|
17
|
Burgess AW. Regulation of Signaling from the Epidermal Growth Factor Family. THE JOURNAL OF PHYSICAL CHEMISTRY C 2022. [DOI: 10.1021/acs.jpcc.2c04156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Antony W. Burgess
- Honorary Laboratory Head, Personalized Oncology Division, WEHI, Parkville 3050, Australia
- Professor Emeritus, Departments of Medical Biology and Surgery (Royal Melbourne Hospital), University of Melbourne, Melbourne 3052, Australia
- The Brain Cancer Centre at WEHI, Parkville 3052, Australia
| |
Collapse
|
18
|
Pellerin È, Pellerin FA, Chabaud S, Pouliot F, Bolduc S, Pelletier M. Bisphenols A and S Alter the Bioenergetics and Behaviours of Normal Urothelial and Bladder Cancer Cells. Cancers (Basel) 2022; 14:cancers14164011. [PMID: 36011004 PMCID: PMC9406715 DOI: 10.3390/cancers14164011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/14/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Bisphenol A (BPA) and bisphenol S (BPS) are used in the production of plastics. These endocrine disruptors can be released into the environment and food, resulting in the continuous exposure of humans to bisphenols (BPs). The bladder urothelium is chronically exposed to BPA and BPS due to their presence in human urine samples. BPA and BPS exposure has been linked to cancer progression, especially for hormone-dependent cancers. However, the bladder is not recognized as a hormone-dependent tissue. Still, the presence of hormone receptors on the urothelium and their role in bladder cancer initiation and progression suggest that BPs could impact bladder cancer development. The effects of chronic exposure to BPA and BPS for 72 h on the bioenergetics (glycolysis and mitochondrial respiration), proliferation and migration of normal urothelial cells and non-invasive and invasive bladder cancer cells were evaluated. The results demonstrate that chronic exposure to BPs decreased urothelial cells' energy metabolism and properties while increasing them for bladder cancer cells. These findings suggest that exposure to BPA and BPS could promote bladder cancer development with a potential clinical impact on bladder cancer progression. Further studies using 3D models would help to understand the clinical consequences of this exposure.
Collapse
Affiliation(s)
- Ève Pellerin
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1J 1Z4, Canada
| | - Félix-Antoine Pellerin
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1J 1Z4, Canada
| | - Stéphane Chabaud
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1J 1Z4, Canada
| | - Frédéric Pouliot
- Oncology Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1R 2J6, Canada
- Department of Surgery, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
| | - Stéphane Bolduc
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1J 1Z4, Canada
- Department of Surgery, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
- Correspondence: (S.B.); (M.P.); Tel.: +1-418-525-4444 (ext. 42282) (S.B.); +1-418-525-4444 (ext. 46166) (M.P.)
| | - Martin Pelletier
- Infectious and Immune Disease Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
- Correspondence: (S.B.); (M.P.); Tel.: +1-418-525-4444 (ext. 42282) (S.B.); +1-418-525-4444 (ext. 46166) (M.P.)
| |
Collapse
|
19
|
Secchi M, Lodola C, Garbelli A, Bione S, Maga G. DEAD-Box RNA Helicases DDX3X and DDX5 as Oncogenes or Oncosuppressors: A Network Perspective. Cancers (Basel) 2022; 14:cancers14153820. [PMID: 35954483 PMCID: PMC9367324 DOI: 10.3390/cancers14153820] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The transformation of a normal cell into a cancerous one is caused by the deregulation of different metabolic pathways, involving a complex network of protein–protein interactions. The cellular enzymes DDX3X and DDX5 play important roles in the maintenance of normal cell metabolism, but their deregulation can accelerate tumor transformation. Both DDX3X and DDX5 interact with hundreds of different cellular proteins, and depending on the specific pathways in which they are involved, both proteins can either act as suppressors of cancer or as oncogenes. In this review, we summarize the current knowledge about the roles of DDX3X and DDX5 in different tumors. In addition, we present a list of interacting proteins and discuss the possible contribution of some of these protein–protein interactions in determining the roles of DDX3X and DDX5 in the process of cancer proliferation, also suggesting novel hypotheses for future studies. Abstract RNA helicases of the DEAD-box family are involved in several metabolic pathways, from transcription and translation to cell proliferation, innate immunity and stress response. Given their multiple roles, it is not surprising that their deregulation or mutation is linked to different pathological conditions, including cancer. However, while in some cases the loss of function of a given DEAD-box helicase promotes tumor transformation, indicating an oncosuppressive role, in other contexts the overexpression of the same enzyme favors cancer progression, thus acting as a typical oncogene. The roles of two well-characterized members of this family, DDX3X and DDX5, as both oncogenes and oncosuppressors have been documented in several cancer types. Understanding the interplay of the different cellular contexts, as defined by the molecular interaction networks of DDX3X and DDX5 in different tumors, with the cancer-specific roles played by these proteins could help to explain their apparently conflicting roles as cancer drivers or suppressors.
Collapse
|
20
|
Akkour K, Alanazi IO, Alfadda AA, Alhalal H, Masood A, Musambil M, Rahman AMA, Alwehaibi MA, Arafah M, Bassi A, Benabdelkamel H. Tissue-Based Proteomic Profiling in Patients with Hyperplasia and Endometrial Cancer. Cells 2022; 11:cells11132119. [PMID: 35805203 PMCID: PMC9265283 DOI: 10.3390/cells11132119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 12/24/2022] Open
Abstract
Uterine cancers are among the most prevalent gynecological malignancies, and endometrial cancer (EC) is the most common in this group. This study used tissue-based proteomic profiling analysis in patients with endometrial cancer and hyperplasia, and control patients. Conventional 2D gel electrophoresis, followed by a mass spectrometry approach with bioinformatics, including a network pathway analysis pipeline, was used to identify differentially expressed proteins and associated metabolic pathways between the study groups. Thirty-six patients (twelve with endometrial cancer, twelve with hyperplasia, and twelve controls) were enrolled in this study. The mean age of the participants was 46–75 years. Eighty-seven proteins were significantly differentially expressed between the study groups, of which fifty-three were significantly differentially regulated (twenty-eight upregulated and twenty-five downregulated) in the tissue samples of EC patients compared to the control (Ctrl). Furthermore, 26 proteins were significantly dysregulated (8 upregulated and 18 downregulated) in tissue samples of hyperplasia (HY) patients compared to Ctrl. Thirty-two proteins (nineteen upregulated and thirteen downregulated) including desmin, peptidyl prolyl cis-trans isomerase A, and zinc finger protein 844 were downregulated in the EC group compared to the HY group. Additionally, fructose bisphosphate aldolase A, alpha enolase, and keratin type 1 cytoskeletal 10 were upregulated in the EC group compared to those in the HY group. The proteins identified in this study were known to regulate cellular processes (36%), followed by biological regulation (16%). Ingenuity pathway analysis found that proteins that are differentially expressed between EC and HY are linked to AKT, ACTA2, and other signaling pathways. The panels of protein markers identified in this study could be used as potential biomarkers for distinguishing between EC and HY and early diagnosis and progression of EC from hyperplasia and normal patients.
Collapse
Affiliation(s)
- Khalid Akkour
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (K.A.); (H.A.); (A.B.)
| | - Ibrahim O. Alanazi
- The National Center for Biotechnology (NCB), Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Assim A. Alfadda
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.A.A.); (A.M.); (M.M.); (M.A.A.)
- Department of Medicine, College of Medicine and King Saud Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Hani Alhalal
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (K.A.); (H.A.); (A.B.)
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.A.A.); (A.M.); (M.M.); (M.A.A.)
| | - Mohthash Musambil
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.A.A.); (A.M.); (M.M.); (M.A.A.)
| | - Anas M. Abdel Rahman
- Metabolomics Section, Department of Clinical Genomics, Center for Genome Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh 11211, Saudi Arabia;
| | - Moudi A. Alwehaibi
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.A.A.); (A.M.); (M.M.); (M.A.A.)
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11461, Saudi Arabia
| | - Maria Arafah
- Department of Pathology, College of Medicine, King Saud University, King Saud University Medical City, Riyadh 11461, Saudi Arabia;
| | - Ali Bassi
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (K.A.); (H.A.); (A.B.)
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.A.A.); (A.M.); (M.M.); (M.A.A.)
- Correspondence:
| |
Collapse
|
21
|
Shu W, Zhu X, Wang K, Cherepanoff S, Conway RM, Madigan MC, Zhu H, Zhu L, Murray M, Zhou F. The multi-kinase inhibitor afatinib serves as a novel candidate for the treatment of human uveal melanoma. Cell Oncol (Dordr) 2022; 45:601-619. [PMID: 35781872 PMCID: PMC9424141 DOI: 10.1007/s13402-022-00686-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2022] [Indexed: 11/03/2022] Open
Abstract
PURPOSE Uveal melanoma (UM) is the most common intraocular malignancy in adults with a poor prognosis and a high recurrence rate. Currently there is no effective treatment for UM. Multi-kinase inhibitors targeting dysregulated pro-tumorigenic signalling pathways have revolutionised anti-cancer treatment but, as yet, their efficacy in UM has not been established. Here, we identified the multi-kinase inhibitor afatinib as a highly effective agent that exerts anti-UM effects in in vitro, ex vivo and in vivo models. METHODS We assessed the anti-cancer effects of afatinib using cell viability, cell death and cell cycle assays in in vitro and ex vivo UM models. The signaling pathways involved in the anti-UM effects of afatinib were evaluated by Western blotting. The in vivo activity of afatinib was evaluated in UM xenograft models using tumour mass measurement, PET scan, immunohistochemical staining and TUNEL assays. RESULTS We found that afatinib reduced cell viability and activated apoptosis and cell cycle arrest in multiple established UM cell lines and in patient tumour-derived primary cell lines. Afatinib impaired cell migration and enhanced reproductive death in these UM cell models. Afatinib-induced cell death was accompanied by activation of STAT1 expression and downregulation of Bcl-xL and cyclin D1 expression, which control cell survival and cell cycle progression. Afatinib attenuated HER2-AKT/ERK/PI3K signalling in UM cell lines. Consistent with these observations, we found that afatinib suppressed tumour growth in UM xenografted mice. CONCLUSION Our data indicate that afatinib activates UM cell death and targets the HER2-mediated cascade, which modulates STAT1-Bcl-xL/cyclin D1 signalling. Thus, targeting HER2 with agents like afatinib may be a novel therapeutic strategy to treat UM and to prevent metastasis.
Collapse
Affiliation(s)
- Wenying Shu
- Department of Pharmacy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 511400, Guangdong Province, China
| | - Xue Zhu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China
| | - Ke Wang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China
| | - Svetlana Cherepanoff
- SydPath, Department of Anatomical Pathology, St Vincent's Hospital, Darlinghurst, NSW, 2010, Australia
| | - R Max Conway
- Ocular Oncology Unit, Sydney Eye Hospital and The Kinghorn Cancer Centre, Sydney, NSW, 2006, Australia.,Save Sight Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Michele C Madigan
- Save Sight Institute, The University of Sydney, Sydney, NSW, 2006, Australia.,School of Optometry and Vision Sciences, University of New South Wales, Sydney, NSW, 2006, Australia
| | - Hong Zhu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
| | - Ling Zhu
- Save Sight Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Michael Murray
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Fanfan Zhou
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
22
|
Varlet P, Bouffet E, Casanova M, Giangaspero F, Antonelli M, Hargrave D, Ladenstein R, Pearson A, Hawkins C, König FB, Rüschoff J, Schmauch C, Bühnemann C, Garin-Chesa P, Schweifer N, Uttenreuther-Fischer M, Gibson N, Ittrich C, Krämer N, Solca F, Stolze B, Geoerger B. Comprehensive analysis of the ErbB receptor family in pediatric nervous system tumors and rhabdomyosarcoma. Pediatr Blood Cancer 2022; 69:e29316. [PMID: 34546642 DOI: 10.1002/pbc.29316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/05/2021] [Accepted: 08/01/2021] [Indexed: 11/11/2022]
Abstract
BACKGROUND There is a paucity of knowledge regarding pediatric biomarkers, including the relevance of ErbB pathway aberrations in pediatric tumors. We investigated the occurrence of ErbB receptor aberrations across different pediatric malignancies, to identify patterns of ErbB dysregulation and define biomarkers suitable for patient enrichment in clinical studies. PROCEDURE Tissue samples from 297 patients with nervous system tumors and rhabdomyosarcoma were analyzed for immunohistochemical expression or gene amplification of epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2). Exploratory analyses of HER3/HER4 expression, and mRNA expression of ErbB receptors/ligands (NanoString) were performed. Assay validation followed general procedures, with additional validation to address Clinical Laboratory Improvement Amendments (CLIA) requirements. RESULTS In most tumor types, samples with high ErbB receptor expression were found with heterogeneous distribution. We considered increased/aberrant ErbB pathway activation when greater than or equal to two EGFR/HER2 markers were simultaneously upregulated. ErbB pathway dysregulation was identified in ∼20%-30% of samples for most tumor types (medulloblastoma/primitive neuroectodermal tumors 31.1%, high-grade glioma 27.1%, neuroblastoma 22.7%, rhabdomyosarcoma 23.1%, ependymoma 18.8%), 4.2% of diffuse intrinsic pontine gliomas, and no recurrent or refractory low-grade astrocytomas. In medulloblastoma/primitive neuroectodermal tumors and neuroblastoma, this was attributed mainly to high EGFR polysomy/HER2 amplification, whereas EGFR gene amplification was observed in some high-grade glioma samples. EGFR/HER2 overexpression was most prevalent in ependymoma. CONCLUSIONS Overexpression and/or amplification of EGFR/HER2 were identified as potential enrichment biomarkers for clinical trials of ErbB-targeted drugs.
Collapse
Affiliation(s)
- Pascale Varlet
- GHU Psychiatrie et Neurosciences, site Sainte-Anne, service de Neuropathologie, Paris, France
| | - Eric Bouffet
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | - Darren Hargrave
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Ruth Ladenstein
- Department of Paediatrics, St. Anna Children's Cancer Research Institute, Medical University, Vienna, Austria
| | - Andy Pearson
- Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK.,Division of Clinical Studies, Institute of Cancer Research, London, UK
| | | | | | | | | | | | - Pilar Garin-Chesa
- Staburo GmbH, Munich, Germany, on behalf of Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Norbert Schweifer
- Staburo GmbH, Munich, Germany, on behalf of Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Neil Gibson
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Carina Ittrich
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Nicole Krämer
- Staburo GmbH, Munich, Germany, on behalf of Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Flavio Solca
- Boehringer Ingelheim RCV GmbH & Co. KG, Vienna, Austria
| | - Britta Stolze
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Birgit Geoerger
- Gustave Roussy Cancer Center, Department of Pediatric and Adolescent Oncology, INSERM U1015, Université Paris Saclay, Villejuif, France
| |
Collapse
|
23
|
Yi X, Zhou Y, Zheng H, Wang L, Xu T, Fu C, Su X. Prognostic targets recognition of rectal adenocarcinoma based on transcriptomics. Medicine (Baltimore) 2021; 100:e25909. [PMID: 34397867 PMCID: PMC8360489 DOI: 10.1097/md.0000000000025909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/22/2021] [Indexed: 01/04/2023] Open
Abstract
Colorectal cancer is currently the third most common cancer around the world. In this study, we chose a bioinformatics analysis method based on network analysis to dig out the pathological mechanism and key prognostic targets of rectal adenocarcinoma (READ).In this study, we downloaded the clinical information data and transcriptome data from the Cancer Genome Atlas database. Differentially expressed genes analysis was used to identify the differential expressed genes in READ. Community discovery algorithm analysis and Correlation analysis between gene modules and clinical data were performed to mine the key modules related to tumor proliferation, metastasis, and invasion. Genetic significance (GS) analysis and PageRank algorithm analysis were applied for find key genes in the key module. Finally, the importance of these genes was confirmed by survival analysis.Transcriptome datasets of 165 cancer tissue samples and 9 paracancerous tissue samples were selected. Gene coexpression networks were constructed, multilevel algorithm was used to divide the gene coexpression network into 11 modules. From GO enrichment analysis, module 11 significantly associated with clinical characteristic N, T, and event, mainly involved in 2 types of biological processes which were highly related to tumor metastasis, invasion, and tumor microenvironment regulation: cell development and differentiation; the development of vascular and nervous systems. Based on the results of survival analysis, 7 key genes were found negatively correlated to the survival rate of READ, such as MMP14, SDC2, LAMC1, ELN, ACTA2, ZNF532, and CYBRD1.Our study found that these key genes were predicted playing an important role in tumor invasion and metastasis, and being associated with the prognosis of READ. This may provide some new potential therapeutic targets and thoughts for the prognosis of READ.
Collapse
Affiliation(s)
- Xingcheng Yi
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Yulai Zhou
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Hanyu Zheng
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Luoying Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Tong Xu
- Jilin Prochance Precision Medicine Experimental Center & Jilin Prochance Biomedical Co., Ltd., Changchun, China
| | - Cong Fu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, and National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyun Su
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| |
Collapse
|
24
|
Mo S, Dai W, Zhou Z, Gu R, Li Y, Xiang W, Han L, Zhang L, Wang R, Cai G, Cai S, Gan L, Li Q. Comprehensive Transcriptomic Analysis Reveals Prognostic Value of an EMT-Related Gene Signature in Colorectal Cancer. Front Cell Dev Biol 2021; 9:681431. [PMID: 34211976 PMCID: PMC8239228 DOI: 10.3389/fcell.2021.681431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/25/2021] [Indexed: 11/16/2022] Open
Abstract
Lymph node metastasis (LNM) is closely related to the postoperative recurrence of colorectal cancer (CRC), and greatly affects patient survival. Conducting Gene set variation analysis (GSVA) and gene set enrichment analysis (GSEA), we found that the epithelial-mesenchymal transition (EMT) signaling pathway is the signaling pathway most relevant to the process of LNM. An EMT-related gene signature was identified from a discovery dataset obtained 489 patients using LIMMA and LASSO Cox methods. Six external independent dataset analyses including a total of 1,045 CRC patients and stratification analysis showed that EMT-related gene signature could sort out those high- and low-risk CRC patients accurately. Functional analysis and loss-of-function exploration in vitro and in vivo indicated that the EMT-related-signature-associated coding genes might play functional roles in the sophisticated regulation of CRC proliferation and metastasis. Prognostic nomograms integrating the EMT-related gene signature and clinicopathological risk factors were constructed for use as numerical prediction tools to assess clinical prognosis and clinical decision-makings. The comprehensive transcriptomic analysis in this article highlights the prognostic value of an EMT-related gene signature for postoperative disease recurrence in CRC patients and reveals a potential prognostic and therapeutic biomarker for CRC.
Collapse
Affiliation(s)
- Shaobo Mo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weixing Dai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zheng Zhou
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruiqi Gu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yaqi Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenqiang Xiang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lingyu Han
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Long Zhang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Renjie Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guoxiang Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Lu Gan
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.,Center of Evidence-Based Medicine, Fudan University, Shanghai, China
| | - Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Wang Y, Li Z, Zhang Z, Chen X. Identification ACTA2 and KDR as key proteins for prognosis of PD-1/PD-L1 blockade therapy in melanoma. Animal Model Exp Med 2021; 4:138-150. [PMID: 34179721 PMCID: PMC8212820 DOI: 10.1002/ame2.12154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
Programmed cell death protein 1 (PD-1) /programmed cell death ligand 1 (PD-L1) blockade is an important therapeutic strategy for melanoma, despite its low clinical response. It is important to identify genes and pathways that may reflect the clinical outcomes of this therapy in patients. We analyzed clinical dataset GSE96619, which contains clinical information from five melanoma patients before and after anti-PD-1 therapy (five pairs of data). We identified 704 DEGs using these five pairs of data, and then the number of DEGs was narrowed down to 286 in patients who responded to treatment. Next, we performed KEGG pathway enrichment and constructed a DEG-associated protein-protein interaction network. Smooth muscle actin 2 (ACTA2) and tyrosine kinase growth factor receptor (KDR) were identified as the hub genes, which were significantly downregulated in the tumor tissue of the two patients who responded to treatment. To confirm our analysis, we demonstrated similar expression tendency to the clinical data for the two hub genes in a B16F10 subcutaneous xenograft model. This study demonstrates that ACTA2 and KDR are valuable responsive markers for PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Yuchen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation StudyInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhaojun Li
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation StudyInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhihui Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation StudyInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation StudyInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
26
|
Suresh R, Diaz RJ. The remodelling of actin composition as a hallmark of cancer. Transl Oncol 2021; 14:101051. [PMID: 33761369 PMCID: PMC8008238 DOI: 10.1016/j.tranon.2021.101051] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
Actin is a key structural protein that makes up the cytoskeleton of cells, and plays a role in functions such as division, migration, and vesicle trafficking. It comprises six different cell-type specific isoforms: ACTA1, ACTA2, ACTB, ACTC1, ACTG1, and ACTG2. Abnormal actin isoform expression has been reported in many cancers, which led us to hypothesize that it may serve as an early biomarker of cancer. We show an overview of the different actin isoforms and highlight mechanisms by which they may contribute to tumorigenicity. Furthermore, we suggest how the aberrant expression of actin subunits can confer cells with greater proliferation ability, increased migratory capability, and chemoresistance through incorporation into the normal cellular F-actin network and altered actin binding protein interaction. Studying this fundamental change that takes place within cancer cells can further our understanding of neoplastic transformation in multiple tissue types, which can ultimately aid in the early-detection, diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Rahul Suresh
- Montreal Neurological Institute, Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | - Roberto J Diaz
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, Faculty of Medicine, McGill University, Montreal, Canada.
| |
Collapse
|
27
|
Ambrosetti E, Bernardinelli G, Hoffecker I, Hartmanis L, Kiriako G, de Marco A, Sandberg R, Högberg B, Teixeira AI. A DNA-nanoassembly-based approach to map membrane protein nanoenvironments. NATURE NANOTECHNOLOGY 2021; 16:85-95. [PMID: 33139936 DOI: 10.1038/s41565-020-00785-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 09/21/2020] [Indexed: 06/11/2023]
Abstract
Most proteins at the plasma membrane are not uniformly distributed but localize to dynamic domains of nanoscale dimensions. To investigate their functional relevance, there is a need for methods that enable comprehensive analysis of the compositions and spatial organizations of membrane protein nanodomains in cell populations. Here we describe the development of a non-microscopy-based method for ensemble analysis of membrane protein nanodomains. The method, termed nanoscale deciphering of membrane protein nanodomains (NanoDeep), is based on the use of DNA nanoassemblies to translate membrane protein organization information into a DNA sequencing readout. Using NanoDeep, we characterized the nanoenvironments of Her2, a membrane receptor of critical relevance in cancer. Importantly, we were able to modulate by design the inventory of proteins analysed by NanoDeep. NanoDeep has the potential to provide new insights into the roles of the composition and spatial organization of protein nanoenvironments in the regulation of membrane protein function.
Collapse
Affiliation(s)
- Elena Ambrosetti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Giulio Bernardinelli
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ian Hoffecker
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Leonard Hartmanis
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Georges Kiriako
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ario de Marco
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Nova Gorica, Slovenia
| | - Rickard Sandberg
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Björn Högberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ana I Teixeira
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
28
|
Amjad E, Asnaashari S, Sokouti B, Dastmalchi S. Systems biology comprehensive analysis on breast cancer for identification of key gene modules and genes associated with TNM-based clinical stages. Sci Rep 2020; 10:10816. [PMID: 32616754 PMCID: PMC7331704 DOI: 10.1038/s41598-020-67643-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Breast cancer (BC), as one of the leading causes of death among women, comprises several subtypes with controversial and poor prognosis. Considering the TNM (tumor, lymph node, metastasis) based classification for staging of breast cancer, it is essential to diagnose the disease at early stages. The present study aims to take advantage of the systems biology approach on genome wide gene expression profiling datasets to identify the potential biomarkers involved at stage I, stage II, stage III, and stage IV as well as in the integrated group. Three HER2-negative breast cancer microarray datasets were retrieved from the GEO database, including normal, stage I, stage II, stage III, and stage IV samples. Additionally, one dataset was also extracted to test the developed predictive models trained on the three datasets. The analysis of gene expression profiles to identify differentially expressed genes (DEGs) was performed after preprocessing and normalization of data. Then, statistically significant prioritized DEGs were used to construct protein-protein interaction networks for the stages for module analysis and biomarker identification. Furthermore, the prioritized DEGs were used to determine the involved GO enrichment and KEGG signaling pathways at various stages of the breast cancer. The recurrence survival rate analysis of the identified gene biomarkers was conducted based on Kaplan-Meier methodology. Furthermore, the identified genes were validated not only by using several classification models but also through screening the experimental literature reports on the target genes. Fourteen (21 genes), nine (17 genes), eight (10 genes), four (7 genes), and six (8 genes) gene modules (total of 53 unique genes out of 63 genes with involving those with the same connectivity degree) were identified for stage I, stage II, stage III, stage IV, and the integrated group. Moreover, SMC4, FN1, FOS, JUN, and KIF11 and RACGAP1 genes with the highest connectivity degrees were in module 1 for abovementioned stages, respectively. The biological processes, cellular components, and molecular functions were demonstrated for outcomes of GO analysis and KEGG pathway assessment. Additionally, the Kaplan-Meier analysis revealed that 33 genes were found to be significant while considering the recurrence-free survival rate as an alternative to overall survival rate. Furthermore, the machine learning calcification models show good performance on the determined biomarkers. Moreover, the literature reports have confirmed all of the identified gene biomarkers for breast cancer. According to the literature evidence, the identified hub genes are highly correlated with HER2-negative breast cancer. The 53-mRNA signature might be a potential gene set for TNM based stages as well as possible therapeutics with potentially good performance in predicting and managing recurrence-free survival rates at stages I, II, III, and IV as well as in the integrated group. Moreover, the identified genes for the TNM-based stages can also be used as mRNA profile signatures to determine the current stage of the breast cancer.
Collapse
Affiliation(s)
- Elham Amjad
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Asnaashari
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Babak Sokouti
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
29
|
Pan F, Wang Q, Li S, Huang R, Wang X, Liao X, Mo H, Zhang L, Zhou X. Prognostic value of key genes of the JAK-STAT signaling pathway in patients with cutaneous melanoma. Oncol Lett 2020; 19:1928-1946. [PMID: 32194688 PMCID: PMC7039088 DOI: 10.3892/ol.2020.11287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 11/22/2019] [Indexed: 01/05/2023] Open
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway is involved in cell immunity, division and death, as well as in tumor formation. The expression of key genes in the JAK-STAT signaling pathway in different types of cancer serves different roles. However, few reports are available on the prognostic value of the genes of the JAK-STAT signaling pathway in skin cutaneous melanoma (SKCM). The potential prognostic value of gene expression in the JAK-STAT signaling pathway in patients with SKCM was analyzed in the present study using data obtained from The Cancer Genome Atlas. To predict the potential functions and mechanisms of these genes in SKCM, gene set enrichment analysis (GSEA) and bioinformatics analysis were performed. A nomogram model including gene expression level and high risk factors was used to predict the risk level of prognostic. High expression levels of STAT1, STAT3, STAT4 and STAT5B, and low expression levels of STAT6 were associated with favorable prognosis [adjusted P<0.001; hazard ratio (HR), 0.595; 95% confidence interval (CI), 0.455–0.778; adjusted P=0.018; HR, 0.725; 95% CI, 0.555–0.947; adjusted P<0.001; HR, 0.590; 95% CI, 0.450–0.773; adjusted P=0.007; HR, 0.690; 95% CI, 0.526–0.940; and adjusted P=0.026; HR, 0.737, 95% CI, 0.563–0.964, respectively]. GSEA results demonstrated that these genes were involved in cell differentiation, invasion, adhesion, migration, cycle, colony formation and mitogen-activated protein kinase signaling. The combination of genes with favorable prognosis had a better effect on the overall survival (univariate survival analysis, P<0.05). The results of the present study suggest that STAT1, STAT3, STAT4, STAT5B and STAT6 gene expression may be used as a potential prognostic biomarker of SKCM, and the combined outcomes may exhibit a stronger interaction and higher survival time for SKCM.
Collapse
Affiliation(s)
- Fuqiang Pan
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Qiaoqi Wang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Sizhu Li
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Rui Huang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Haiyan Mo
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Liming Zhang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Xiang Zhou
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| |
Collapse
|
30
|
Timmermans-Sprang EPM, Mestemaker HM, Steenlage RR, Mol JA. Dasatinib inhibition of cSRC prevents the migration and metastasis of canine mammary cancer cells with enhanced Wnt and HER signalling. Vet Comp Oncol 2019; 17:413-426. [PMID: 31069942 DOI: 10.1111/vco.12490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/05/2018] [Accepted: 04/24/2019] [Indexed: 12/16/2022]
Abstract
Human epidermal growth factor 2 (HER2) overexpression leads to aggressive mammary tumour growth. Although the prognosis of HER2+ tumours in humans is greatly improved using biologicals, therapy resistance, which may be caused by increased phosphatidyl-3-kinase (PI3K), rous sarcoma proto-oncogene (cSRC) or wingless-type MMTV integration site family (Wnt) activity, is a major concern. A recent analysis of 12 canine mammary cell lines showed an association between HER2/3 overexpression and phosphatase and tensin homologue (PTEN) deletion with elevated Wnt-signalling. Wnt-activity appeared to be insensitive to phosphatidyl-3-kinase (PI3K) inhibitors but sensitive to Src-I1. We hypothesized that Wnt activation, was caused by HER2/3-activated cSRC activation. The role of HER2/3 on Wnt signalling was investigated by silencing HER2/3 expression using specific small interfering RNA (siRNAs). Next, the effect of an epidermal growth factor receptor (EGFR)/HER2 tyrosine kinase inhibitor on Wnt activity and migration was investigated and compared to other tyrosine kinase inhibitors (TKIs) of related signalling pathways. Finally, two TKIs, a cSRC and a PI3K inhibitor, were investigated in a zebrafish xenograft model. Silencing of HER1-3 did not inhibit the intrinsic high Wnt activity, whereas the HER kinase inhibitor afatinib showed enhanced Wnt activity. The strongest inhibition of Wnt activity and cell viability and migration was shown by cSRC inhibitors, which also showed strong inhibition of cell viability and metastasis in a zebrafish xenograft model. HER2/3 overexpression or HER2/3-induced cSRC activation is not the cause of enhanced Wnt activity. However, inhibition of cSRC resulted in a strong inhibition of Wnt activity and cell migration and metastasis. Further studies are needed to unravel the mechanism of cSRC activation and cSRC inhibition to restore sensitivity to HER-inhibitors in HER2/3-positive breast cancer.
Collapse
Affiliation(s)
| | - Helena M Mestemaker
- Department of Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
| | - Renske R Steenlage
- Department of Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
| | - Jan A Mol
- Department of Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
31
|
Ju Q, Zhao YJ, Dong Y, Cheng C, Zhang S, Yang Y, Li P, Ge D, Sun B. Identification of a miRNA-mRNA network associated with lymph node metastasis in colorectal cancer. Oncol Lett 2019; 18:1179-1188. [PMID: 31423178 PMCID: PMC6607389 DOI: 10.3892/ol.2019.10460] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 05/07/2019] [Indexed: 02/06/2023] Open
Abstract
Lymph node metastasis is an important step in the progression of colorectal cancer (CRC); however, the underlying mechanisms are still unknown. The aim of the present study was to identify the gene expression pattern during lymph node metastasis in CRC and to identify upstream microRNAs (miRNAs) to explore the underlying mechanisms in detail. A total of 305 differently expressed genes (DEGs) were identified, including 227 upregulated genes and 78 downregulated genes in lymph node metastasis. Pathway and process enrichment analysis demonstrated that DEGs were significantly enriched in ‘NABA CORE MATRISOME’, ‘extracellular matrix assembly’, ‘antimicrobial humoral response’ and ‘Toll-like receptor signaling’ pathways. The top 10 hub genes were identified by protein-protein interaction network, and sub-networks revealed that these genes were involved in significant pathways, including ‘neutrophil chemotaxis’ and ‘Smooth Muscle Contraction’. In addition, 73 mature differently expressed miRNAs associated with lymph node metastasis were identified, of which 48 were upregulated and 25 were downregulated. Six miRNAs were identified to regulate DEGs. Additionally, based on the relationship between miRNAs and transcription factors, a miRNA-TF-mRNA network was constructed. In conclusion, DEGs, miRNAs and their interactions and pathways were identified in lymph node metastasis in CRC, which provided insight into the mechanism of CRC metastasis and may be used to develop novel targets for CRC treatment.
Collapse
Affiliation(s)
- Qiang Ju
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yan-Jie Zhao
- School of Public Health, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yong Dong
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Cong Cheng
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Shaoqiang Zhang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yuanming Yang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Ping Li
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Dongmei Ge
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Bo Sun
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
32
|
Chai D, Li K, Du H, Yang S, Yang R, Xu Y, Lian X. β2-microglobulin has a different regulatory molecular mechanism between ER + and ER - breast cancer with HER2 . BMC Cancer 2019; 19:223. [PMID: 30866857 PMCID: PMC6417228 DOI: 10.1186/s12885-019-5410-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/26/2019] [Indexed: 12/31/2022] Open
Abstract
Background Previous studies have demonstrated that β2-microglobulin (β2M) promotes the growth and survival of a variety of cancer cells and has different regulatory effects on the expression of Bcl-2 and HER2 in HER2− breast cancer cells. However, β2M-mediated signaling in ER+ and ER− breast cancer with HER2− remains unclear. Methods β2M expression vector and siRNA were transfected into two types of HER2− breast cancer cells, and the possible relevant signaling molecules were subsequently analyzed by real-time PCR and western blotting. These signaling molecules were also analyzed by real-time PCR and immunohistochemistry (IHC) in two types of HER2− breast cancer tissues, and the associations between β2M and these signaling molecules were assessed using Spearman’s correlation analysis. Results β2M silencing downregulated p-SGK1/SGK1 levels and Bcl-2 expression, and β2M overexpression downregulated p-CREB/CREB and significantly upregulated p-SGK1/SGK1 levels and Bcl-2 expression, and both resulting processes did not affect HER2, HIF-1α, VEGF, and ERK signaling in ER+ breast cancer cells with HER2−. β2M silencing upregulated p-CREB/CREB and VEGF protein and significantly downregulated p-ERK/ERK levels, and β2M overexpression downregulated p-CREB/CREB and VEGF, significantly upregulated p-ERK/ERK levels, and both resulting processes did not affect HIF-1α and SGK1 signaling in ER− breast cancer cells with HER2−. β2M expression was positively correlated with p-CREB, p-SGK1, and Bcl-2 expression and had no correlation with HIF-1α, VEGF, and p-ERK1/2, whereas p-SGK1 exhibited a significantly positive correlation with Bcl-2 expression in cancer tissues of patients with luminal A breast cancer, which coincide with the results obtained from the same molecular types of breast cancer cells except CREB signaling. However, β2M expression did not show a significant correlation with HIF-1α, p-CREB, VEGF, p-SGK1, p-ERK1/2, and Bcl-2 expression in cancer tissues of patients with basal-like breast cancer, which was discordant with the results obtained from the same molecular types of breast cancer cells. Conclusions β2M has a different molecular regulatory mechanism between ER+ and ER− breast cancer with HER2−, and it may promote tumor survival through the SGK1/Bcl-2 signaling pathway in ER+ breast cancer with HER2− and has no regulatory effects on ER− breast cancer with HER2−.
Collapse
Affiliation(s)
- Dandan Chai
- Department of Medicine Biotechnology, Medicine and Science Research Institute of Gansu Province, Lanzhou, China
| | - Kesheng Li
- Department of Medicine Biotechnology, Medicine and Science Research Institute of Gansu Province, Lanzhou, China.
| | - Huifen Du
- Department of Medicine Biotechnology, Medicine and Science Research Institute of Gansu Province, Lanzhou, China
| | - Suisheng Yang
- Department of Breast Surgery, Tumor Hospital of Gansu Province, Lanzhou, China
| | - Rong Yang
- Department of Pathology, Tumor Hospital of Gansu Province, Lanzhou, China
| | - Yang Xu
- Department of Medicine Biotechnology, Medicine and Science Research Institute of Gansu Province, Lanzhou, China
| | - Xiaowen Lian
- Department of Medicine Biotechnology, Medicine and Science Research Institute of Gansu Province, Lanzhou, China
| |
Collapse
|
33
|
Targeting of JAK-STAT Signaling in Breast Cancer: Therapeutic Strategies to Overcome Drug Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1152:271-281. [PMID: 31456189 DOI: 10.1007/978-3-030-20301-6_14] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Rapidly emerging ground-breaking discoveries have provided near to complete resolution of breast cancer signaling landscape and scientists have mapped the knowledge gaps associated with proteins encoded by the human genome. Based on the insights gleaned from decades of research, it seems clear that ligands transmit distinct information through specific receptors that is processed into characteristically unique biological outputs. Advances in imaging, structural biology, proteomics and genome editing have helped us to gain new insights into JAK-STAT signaling and how alterations in this pathway contributed to development of breast cancer and metastatic spread. Data obtained through high-throughput technologies has started to shed light on signal-transducer complexes formed during JAK-STAT signaling. Pharmacologists and molecular biologists are focusing on the strategies to therapeutically target this pathway to overcome drug resistance associated with breast cancer.
Collapse
|
34
|
Lin J, Qi W, Chen K, Yan Y, Li X, Feng Z, Pan X. Downregulating STAT1/caspase‐3 signaling with fludarabine to alleviate progression in a rat model of steroid‐induced avascular necrosis of the femoral head. J Biochem Mol Toxicol 2018; 33:e22265. [PMID: 30506661 DOI: 10.1002/jbt.22265] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/03/2018] [Accepted: 10/24/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Jian Lin
- Department of OrthopaedicThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou China
- Key Laboratory of Orthopaedics of Zhejiang ProvinceWenzhou China
- The Second School of Medicine, Wenzhou Medical UniversityWenzhou China
| | - Weihui Qi
- Department of OrthopaedicThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou China
- Key Laboratory of Orthopaedics of Zhejiang ProvinceWenzhou China
- The Second School of Medicine, Wenzhou Medical UniversityWenzhou China
| | - Kai Chen
- Department of OrthopaedicThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou China
- Key Laboratory of Orthopaedics of Zhejiang ProvinceWenzhou China
- The Second School of Medicine, Wenzhou Medical UniversityWenzhou China
| | - Yingzhao Yan
- Department of OrthopaedicThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou China
- Key Laboratory of Orthopaedics of Zhejiang ProvinceWenzhou China
- The Second School of Medicine, Wenzhou Medical UniversityWenzhou China
| | - Xiaobin Li
- Department of OrthopaedicThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou China
- Key Laboratory of Orthopaedics of Zhejiang ProvinceWenzhou China
- The Second School of Medicine, Wenzhou Medical UniversityWenzhou China
| | - Zhenhua Feng
- Department of Orthopaedic SurgeryThe Second Affiliated Hospital of Jiaxing UniversityJiaxing China
| | - Xiaoyun Pan
- Department of OrthopaedicThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou China
- Key Laboratory of Orthopaedics of Zhejiang ProvinceWenzhou China
- The Second School of Medicine, Wenzhou Medical UniversityWenzhou China
| |
Collapse
|
35
|
Sagara Y, Takada M, Ohi Y, Ohtani S, Kurozumi S, Inoue K, Kosaka Y, Hattori M, Yamashita T, Takao S, Sato N, Iwata H, Kurosumi M, Toi M. Effectiveness of neo-adjuvant systemic therapy with trastuzumab for basal HER2 type breast cancer: results from retrospective cohort study of Japan Breast Cancer Research Group (JBCRG)-C03. Breast Cancer Res Treat 2018; 171:675-683. [PMID: 29971625 DOI: 10.1007/s10549-018-4873-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/29/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE While human epidermal growth factor receptor 2 (HER2) target therapies have significantly improved the prognosis of patients with HER2-enriched breast cancer, differing clinical benefits and gene expression analyses suggest a divergent HER2 subgroup. We aimed to investigate whether the basal HER2 subtype of breast cancer has distinguished characteristics. METHODS We performed a substudy by using data from a retrospective multi-institutional cohort of JBCRG-C03. Between 2001 and 2011, we identified 184 eligible patients who received concurrent neo-adjuvant chemotherapy (NAC) with trastuzumab for hormone receptor-negative and HER2-positive breast cancer. We defined basal HER2 subtype breast cancer as HER2-positive, ER/PgR-negative, and basal markers (EGFR, CK14 or CK5/6) positive by immunohistochemistrical evaluation. The pathologic complete response (pCR) and disease-free survival (DFS) rates were compared between the two subtypes. RESULTS A total of 127 (69.0%) patients achieved pCR after NAC and 29 (15.8%) patients experienced events of DFS within a 42 month median follow-up period (interquartile range 26-58 months). Although the basal HER2 subtype was related with poor DFS (3 year DFS: non-basal HER2, 95.0%; basal HER2, 86.9%; adjusted HR 3.4; 95% CI 1.2-14.5), neither the subtype (pCR: non-basal HER2, 75%; basal HER2, 66.7%; adjusted OR 0.60; 95% CI 0.27-1.28) nor the degree of expression of basal markers was significantly related with the pCR rate. CONCLUSION Basal HER2 phenotype showed poor DFS, but equivalent pCR rate after concurrent neo-adjuvant chemotherapy with trastuzumab. A different treatment approach to basal-HER2 type is needed even for cases that achieved adequate clinical response after NAC.
Collapse
Affiliation(s)
- Yasuaki Sagara
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, 54 Kawaracho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan. .,Department of Breast Surgical Oncology, Social Medical Cooperation Hakuaikai, Kagoshima, Japan.
| | - Masahiro Takada
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, 54 Kawaracho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasuyo Ohi
- Department of Pathology, Social Medical Cooperation Hakuaikai, Kagoshima, Japan
| | - Shoichiro Ohtani
- Department of Breast Surgery, Hiroshima City Hospital, Hiroshima, Japan
| | - Sasagu Kurozumi
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Kenichi Inoue
- Department of Breast Oncology, Saitama Cancer Center, Saitama, Japan
| | - Yoshimasa Kosaka
- Department of Breast and Endocrine Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Masaya Hattori
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Toshinari Yamashita
- Department of Breast and Endocrine Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Shintaro Takao
- Department of Breast Surgery, Hyogo Cancer Center, Akashi, Japan
| | - Nobuaki Sato
- Department of Breast Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | | | - Masakazu Toi
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, 54 Kawaracho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
36
|
Satgé D. A Tumor Profile in Primary Immune Deficiencies Challenges the Cancer Immune Surveillance Concept. Front Immunol 2018; 9:1149. [PMID: 29881389 PMCID: PMC5976747 DOI: 10.3389/fimmu.2018.01149] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/07/2018] [Indexed: 01/23/2023] Open
Abstract
Under the concept of cancer immune surveillance, individuals with primary immune deficiencies would be expected to develop many more malignancies and show an excess of all types of cancers, compared to people with a normal immune system. A review of the nine most frequent and best-documented human conditions with primary immune deficiency reveals a 1.6- to 2.3-fold global increase of cancer in the largest epidemiological studies. However, the spectrum of cancer types with higher frequencies is narrow, limited mainly to lymphoma, digestive tract cancers, and virus-induced cancers. Increased lymphoma is also reported in animal models of immune deficiency. Overstimulation of leukocytes, chronic inflammation, and viruses explain this tumor profile. This raises the question of cancers being foreign organisms or tissues. Organisms, such as bacteria, viruses, and parasites as well as non-compatible grafts are seen as foreign (non-self) and identified and destroyed or rejected by the body (self). As cancer cells rarely show strong (and unique) surface antibodies, their recognition and elimination by the immune system is theoretically questionable, challenging the immune surveillance concept. In the neonatal period, the immune system is weak, but spontaneous regression and good outcomes occur for some cancers, suggesting that non-immune factors are effective in controlling cancer. The idea of cancer as a group of cells that must be destroyed and eliminated appears instead as a legacy of methods and paradigms in microbiological medicine. As an alternative approach, cancer cells could be considered part of the body and could be controlled by an embryonic and neonatal environment.
Collapse
Affiliation(s)
- Daniel Satgé
- Institut Universitaire de Recherche Clinique, Biostatistics, Epidemiology and Public Health, Team Cancer EA 2415 and Oncodéfi, Montpellier, France
| |
Collapse
|
37
|
Wu HT, Liu J, Li GW, Shen JX, Huang YT. The transcriptional STAT3 is a potential target, whereas transcriptional STAT5A/5B/6 are new biomarkers for prognosis in human breast carcinoma. Oncotarget 2017; 8:36279-36288. [PMID: 28422733 PMCID: PMC5482654 DOI: 10.18632/oncotarget.16748] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/21/2017] [Indexed: 02/07/2023] Open
Abstract
Signal Transducer and Activators of Transcription (STAT) is a set of transcription factors, involved in diverse cellular functions. Evidences from cell lines, mouse models and human tissues implicate these transcription factors in the oncogenesis of breast cancer. However, the diverse expression patterns and prognostic values of 7 STATs remain to be elucidated. In the current study, we mined the transcriptional and survival data of STATs in patients with breast carcinoma (BC) through ONCOMINE, bc-GenExMiner, Kaplan-Meier Plotter and cBioPortal. It was found that STAT1/2 were up-regulated, whereas STAT3/4/5A/5B were down-regulated in BC patients compared with the normal tissues. The expressions of STAT5A/5B/6 were correlated with decreased levels of histological differentiation. In survival analyses through the Kaplan-Meier plotter database, high transcription levels of STAT2/4/5A/5B/6 were associated with better relapse-free survival (RFS) in all BC patients. Conversely, high STAT3 predicted shorter RFS in BC patients, suggesting that STAT3 is potential targets for precision therapy to BC patients. These data also provided STAT5A/5B/6 as new biomarker for BC prognosis.
Collapse
Affiliation(s)
- Hua-Tao Wu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, PR China
| | - Jing Liu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, PR China
- Chang Jiang Scholar's Laboratory, Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Shantou University Medical College, Shantou, PR China
| | - Guan-Wu Li
- Open Laboratory for Tumor Molecular Biology, Department of Biochemistry, The Key Lab of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, PR China
| | - Jia-Xin Shen
- Department of Hematology, The First Affiliated Hospital of Shantou University Medical College, Shantou, PR China
| | - Yi-Teng Huang
- Health Care Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, PR China
| |
Collapse
|
38
|
García-Ledo L, Nuevo-Tapioles C, Cuevas-Martín C, Martínez-Reyes I, Soldevilla B, González-Llorente L, Cuezva JM. Overexpression of the ATPase Inhibitory Factor 1 Favors a Non-metastatic Phenotype in Breast Cancer. Front Oncol 2017; 7:69. [PMID: 28443245 PMCID: PMC5385467 DOI: 10.3389/fonc.2017.00069] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/27/2017] [Indexed: 01/03/2023] Open
Abstract
Partial suppression of mitochondrial oxidative phosphorylation and the concurrent activation of aerobic glycolysis is a hallmark of proliferating cancer cells. Overexpression of the ATPase inhibitory factor 1 (IF1), an in vivo inhibitor of the mitochondrial ATP synthase, is observed in most prevalent human carcinomas favoring metabolic rewiring to an enhanced glycolysis and cancer progression. Consistently, a high expression of IF1 in hepatocarcinomas and in carcinomas of the lung, bladder, and stomach and in gliomas is a biomarker of bad patient prognosis. In contrast to these findings, we have previously reported that a high expression level of IF1 in breast carcinomas is indicative of less chance to develop metastatic disease. This finding is especially relevant in the bad prognosis group of patients bearing triple-negative breast carcinomas. To investigate the molecular mechanisms that underlie the differential behavior of IF1 in breast cancer progression, we have developed the triple-negative BT549 breast cancer cell line that overexpresses IF1 stably. When compared to controls, IF1-cells partially shut down respiration and enhance aerobic glycolysis. Transcriptomic analysis suggested that migration and invasion were specifically inhibited in IF1-overexpressing breast cancer cells. Analysis of gene expression by qPCR and western blotting indicate that IF1 overexpression supports the maintenance of components of the extracellular matrix (ECM) and E-cadherin concurrently with the downregulation of components and signaling pathways involved in epithelial to mesenchymal transition. The overexpression of IF1 in breast cancer cells has no effect in the rates of cellular proliferation and in the cell death response to staurosporine and hydrogen peroxide. However, the overexpression of IF1 significantly diminishes the ability of the cells to grow in soft agar and to migrate and invade when compared to control cells. Overall, the results indicate that IF1 overexpression despite favoring a metabolic phenotype prone to cancer progression in the specific case of breast cancer cells also promotes the maintenance of the ECM impeding metastatic disease. These findings hence provide a mechanistic explanation to the better prognosis of breast cancer patients bearing tumors with high expression level of IF1.
Collapse
Affiliation(s)
- Lucía García-Ledo
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristina Nuevo-Tapioles
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Cuevas-Martín
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Inmaculada Martínez-Reyes
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Beatriz Soldevilla
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lucía González-Llorente
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|