1
|
Liermann-Wooldrik KT, Kosmacek EA, Oberley-Deegan RE. Adipose Tissues Have Been Overlooked as Players in Prostate Cancer Progression. Int J Mol Sci 2024; 25:12137. [PMID: 39596205 PMCID: PMC11594286 DOI: 10.3390/ijms252212137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity is a common risk factor in multiple tumor types, including prostate cancer. Obesity has been associated with driving metastasis, therapeutic resistance, and increased mortality. The effect of adipose tissue on the tumor microenvironment is still poorly understood. This review aims to highlight the work conducted in the field of obesity and prostate cancer and bring attention to areas where more research is needed. In this review, we have described key differences between healthy adipose tissues and obese adipose tissues, as they relate to the tumor microenvironment, focusing on mechanisms related to metabolic changes, abnormal adipokine secretion, altered immune cell presence, and heightened oxidative stress as drivers of prostate cancer formation and progression. Interestingly, common treatment options for prostate cancer ignore the adipose tissue located near the site of the tumor. Because of this, we have outlined how excess adipose tissue potentially affects therapeutics' efficacy, such as androgen deprivation, chemotherapy, and radiation treatment, and identified possible drug targets to increase prostate cancer responsiveness to clinical treatments. Understanding how obesity affects the tumor microenvironment will pave the way for understanding why some prostate cancers become metastatic or treatment-resistant, and why patients experience recurrence.
Collapse
Affiliation(s)
| | | | - Rebecca E. Oberley-Deegan
- Department of Biochemistry and Molecular Biology, 985870 University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.T.L.-W.)
| |
Collapse
|
2
|
Xin Z, Qin L, Tang Y, Guo S, Li F, Fang Y, Li G, Yao Y, Zheng B, Zhang B, Wu D, Xiao J, Ni C, Wei Q, Zhang T. Immune mediated support of metastasis: Implication for bone invasion. Cancer Commun (Lond) 2024; 44:967-991. [PMID: 39003618 PMCID: PMC11492328 DOI: 10.1002/cac2.12584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 07/15/2024] Open
Abstract
Bone is a common organ affected by metastasis in various advanced cancers, including lung, breast, prostate, colorectal, and melanoma. Once a patient is diagnosed with bone metastasis, the patient's quality of life and overall survival are significantly reduced owing to a wide range of morbidities and the increasing difficulty of treatment. Many studies have shown that bone metastasis is closely related to bone microenvironment, especially bone immune microenvironment. However, the effects of various immune cells in the bone microenvironment on bone metastasis remain unclear. Here, we described the changes in various immune cells during bone metastasis and discussed their related mechanisms. Osteoblasts, adipocytes, and other non-immune cells closely related to bone metastasis were also included. This review also summarized the existing treatment methods and potential therapeutic targets, and provided insights for future studies of cancer bone metastasis.
Collapse
Affiliation(s)
- Zengfeng Xin
- Department of Orthopedic SurgerySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Luying Qin
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yang Tang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Siyu Guo
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Fangfang Li
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yuan Fang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Gege Li
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yihan Yao
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Binbin Zheng
- Department of Respiratory MedicineNingbo Hangzhou Bay HospitalNingboZhejiangP. R. China
| | - Bicheng Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Dang Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Jie Xiao
- Department of Orthopedic SurgerySecond Affiliated Hospital (Jiande Branch)Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Chao Ni
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Breast SurgerySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Qichun Wei
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Ting Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| |
Collapse
|
3
|
Jung H, Paust S. Chemokines in the tumor microenvironment: implications for lung cancer and immunotherapy. Front Immunol 2024; 15:1443366. [PMID: 39114657 PMCID: PMC11304008 DOI: 10.3389/fimmu.2024.1443366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
The tumor microenvironment (TME) is a complex interconnected network of immune cells, fibroblasts, blood vessels, and extracellular matrix surrounding the tumor. Because of its immunosuppressive nature, the TME can pose a challenge for cancer immunotherapies targeting solid tumors. Chemokines have emerged as a crucial element in enhancing the efficacy of cancer immunotherapy, playing a direct role in immune cell signaling within the TME and facilitating immune cell migration towards cancer cells. However, chemokine ligands and their receptors exhibit context-dependent diversity, necessitating evaluation of their tumor-promoting or inhibitory effects based on tumor type and immune cell characteristics. This review explores the role of chemokines in tumor immunity and metastasis in the context of the TME. We also discuss current chemokine-related advances in cancer immunotherapy research, with a particular focus on lung cancer, a common cancer with a low survival rate and limited immunotherapy options.
Collapse
Affiliation(s)
| | - Silke Paust
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| |
Collapse
|
4
|
de Paula CP, de Oliveira da Silva JPM, Romanello KS, Bernardo VS, Torres FF, da Silva DGH, da Cunha AF. Peroxiredoxins in erythrocytes: far beyond the antioxidant role. J Mol Med (Berl) 2023; 101:1335-1353. [PMID: 37728644 DOI: 10.1007/s00109-023-02368-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/17/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023]
Abstract
The red blood cells (RBCs) are essential to transport oxygen (O2) and nutrients throughout the human body. Changes in the structure or functioning of the erythrocytes can lead to several deficiencies, such as hemolytic anemias, in which an increase in reactive oxidative species generation is involved in the pathophysiological process, playing a significant role in the severity of several clinical manifestations. There are important lines of defense against the damage caused by oxidizing molecules. Among the antioxidant molecules, the enzyme peroxiredoxin (Prx) has the higher decomposition power of hydrogen peroxide, especially in RBCs, standing out because of its abundance. This review aimed to present the recent findings that broke some paradigms regarding the three isoforms of Prxs found in RBC (Prx1, Prx2, and Prx6), showing that in addition to their antioxidant activity, these enzymes may have supplementary roles in transducing peroxide signals, as molecular chaperones, protecting from membrane damage, and maintenance of iron homeostasis, thus contributing to the overall survival of human RBCs, roles that seen to be disrupted in hemolytic anemia conditions.
Collapse
Affiliation(s)
- Carla Peres de Paula
- Genetics and Evolution Department, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil.
- Biotechnology Graduate Program, Exact and Technology Sciences Center, Federal University of São Carlos, São Carlos, Brazil.
| | - João Pedro Maia de Oliveira da Silva
- Genetics and Evolution Department, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil
- Evolutionary Genetics and Molecular Biology Graduate Program, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil
| | - Karen Simone Romanello
- Genetics and Evolution Department, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil
- Evolutionary Genetics and Molecular Biology Graduate Program, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil
| | | | | | - Danilo Grünig Humberto da Silva
- Department of Biology, Paulista State University, São Paulo, Brazil
- Federal University of Mato Grosso do Sul, Campus de Três Lagoas, Três Lagoas, Mato Grosso do Sul, Brazil
| | - Anderson Ferreira da Cunha
- Genetics and Evolution Department, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil.
| |
Collapse
|
5
|
Uchendu I, Zhilenkova A, Pirogova Y, Basova M, Bagmet L, Kohanovskaia I, Ngaha Y, Ikebunwa O, Sekacheva M. Cytokines as Potential Therapeutic Targets and their Role in the Diagnosis and Prediction of Cancers. Curr Pharm Des 2023; 29:2552-2567. [PMID: 37916493 DOI: 10.2174/0113816128268111231024054240] [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: 06/19/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023]
Abstract
The death rate from cancer is declining as a result of earlier identification and more advanced treatments. Nevertheless, a number of unfavourable adverse effects, including prolonged, long-lasting inflammation and reduced immune function, usually coexist with anti-cancer therapies and lead to a general decline in quality of life. Improvements in standardized comprehensive therapy and early identification of a variety of aggressive tumors remain the main objectives of cancer research. Tumor markers in those with cancer are tumor- associated proteins that are clinically significant. Even while several tumor markers are routinely used, they don't always provide reliable diagnostic information. Serum cytokines are promising markers of tumor stage, prognosis, and responsiveness to therapy. In fact, several cytokines are currently proposed as potential biomarkers in a variety of cancers. It has actually been proposed that the study of circulatory cytokines together with biomarkers that are particular to cancer can enhance and accelerate cancer diagnosis and prediction, particularly via blood samples that require minimal to the absence of invasion. The purpose of this review was to critically examine relevant primary research literature in order to elucidate the role and importance of a few identified serum cytokines as prospective therapeutic targets in oncological diseases.
Collapse
Affiliation(s)
- Ikenna Uchendu
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, University of Nigeria, Enugu Campus, Enugu, Nigeria
| | - Angelina Zhilenkova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Yuliya Pirogova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Maria Basova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Leonid Bagmet
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Iana Kohanovskaia
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Yvan Ngaha
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Obinna Ikebunwa
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, University of Nigeria, Enugu Campus, Enugu, Nigeria
- Department of Biotechnology, First Moscow State Medical University of The Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Marina Sekacheva
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| |
Collapse
|
6
|
Fontvieille E, His M, Biessy C, Navionis AS, Torres-Mejía G, Ángeles-Llerenas A, Alvarado-Cabrero I, Sánchez GI, Navarro E, Cortes YR, Porras C, Rodriguez AC, Garmendia ML, Soto JL, Moyano L, Porter PL, Lin MG, Guenthoer J, Romieu I, Rinaldi S. Inflammatory biomarkers and risk of breast cancer among young women in Latin America: a case-control study. BMC Cancer 2022; 22:877. [PMID: 35948877 PMCID: PMC9367082 DOI: 10.1186/s12885-022-09975-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/02/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Breast cancer incidence is increasing rapidly in Latin America, with a higher proportion of cases among young women than in developed countries. Studies have linked inflammation to breast cancer development, but data is limited in premenopausal women, especially in Latin America. METHODS We investigated the associations between serum biomarkers of chronic inflammation (interleukin (IL)-6, IL-8, IL-10, tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), leptin, adiponectin) and risk of premenopausal breast cancer among 453 cases and 453 matched, population-based controls from Chile, Colombia, Costa Rica, and Mexico. Odds ratios (OR) were estimated using conditional logistic regression models. Analyses were stratified by size and hormonal receptor status of the tumors. RESULTS IL-6 (ORper standard deviation (SD) = 1.33 (1.11-1.60)) and TNF-α (ORper SD = 1.32 (1.11-1.58)) were positively associated with breast cancer risk in fully adjusted models. Evidence of heterogeneity by estrogen receptor (ER) status was observed for IL-8 (P-homogeneity = 0.05), with a positive association in ER-negative tumors only. IL-8 (P-homogeneity = 0.06) and TNF-α (P-homogeneity = 0.003) were positively associated with risk in the largest tumors, while for leptin (P-homogeneity = 0.003) a positive association was observed for the smallest tumors only. CONCLUSIONS The results of this study support the implication of chronic inflammation in breast cancer risk in young women in Latin America. Largest studies of prospective design are needed to confirm these findings in premenopausal women.
Collapse
Affiliation(s)
- Emma Fontvieille
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, Lyon, France
| | - Mathilde His
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, Lyon, France
| | - Carine Biessy
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, Lyon, France
| | - Anne-Sophie Navionis
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, Lyon, France
| | - Gabriela Torres-Mejía
- Centre for Population Health Research, National Institute of Public Health, Cuernavaca, Mexico
| | | | - Isabel Alvarado-Cabrero
- Servicio de Patología, Hospital de Oncología, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Gloria Inés Sánchez
- Group Infection and Cancer, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Edgar Navarro
- Grupo Proyecto UNI-Barranquilla, Universidad del Norte, Barranquilla, Colombia
| | | | - Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas (ACIB)-Fundación INCIENSA, San Jose, Costa Rica
| | - Ana Cecilia Rodriguez
- Agencia Costarricense de Investigaciones Biomédicas (ACIB)-Fundación INCIENSA, San Jose, Costa Rica
| | - Maria Luisa Garmendia
- Instituto de Nutrición y de Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | | | | | - Peggy L Porter
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Ming Gang Lin
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Jamie Guenthoer
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Isabelle Romieu
- Centre for Population Health Research, National Institute of Public Health, Cuernavaca, Mexico
- Hubert Department of Global Health, Emory University, Atlanta, Georgia, USA
| | - Sabina Rinaldi
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, Lyon, France.
| |
Collapse
|
7
|
Santolla MF, Talia M, Cirillo F, Scordamaglia D, De Rosis S, Spinelli A, Miglietta AM, Nardo B, Filippelli G, De Francesco EM, Belfiore A, Lappano R, Maggiolini M. The AGEs/RAGE Transduction Signaling Prompts IL-8/CXCR1/2-Mediated Interaction between Cancer-Associated Fibroblasts (CAFs) and Breast Cancer Cells. Cells 2022; 11:2402. [PMID: 35954247 PMCID: PMC9368521 DOI: 10.3390/cells11152402] [Citation(s) in RCA: 6] [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: 07/01/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 01/27/2023] Open
Abstract
Advanced glycation end products (AGEs) and the cognate receptor, named RAGE, are involved in metabolic disorders characterized by hyperglycemia, type 2 diabetes mellitus (T2DM) and obesity. Moreover, the AGEs/RAGE transduction pathway prompts a dysfunctional interaction between breast cancer cells and tumor stroma toward the acquisition of malignant features. However, the action of the AGEs/RAGE axis in the main players of the tumor microenvironment, named breast cancer-associated fibroblasts (CAFs), remains to be fully explored. In the present study, by chemokine array, we first assessed that interleukin-8 (IL-8) is the most up-regulated pro-inflammatory chemokine upon AGEs/RAGE activation in primary CAFs, obtained from breast tumors. Thereafter, we ascertained that the AGEs/RAGE signaling promotes a network cascade in CAFs, leading to the c-Fos-dependent regulation of IL-8. Next, using a conditioned medium from AGEs-exposed CAFs, we determined that IL-8/CXCR1/2 paracrine activation induces the acquisition of migratory and invasive features in MDA-MB-231 breast cancer cells. Altogether, our data provide new insights on the involvement of IL-8 in the AGEs/RAGE transduction pathway among the intricate connections linking breast cancer cells to the surrounding stroma. Hence, our findings may pave the way for further investigations to define the role of IL-8 as useful target for the better management of breast cancer patients exhibiting metabolic disorders.
Collapse
Affiliation(s)
- Maria Francesca Santolla
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Marianna Talia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Domenica Scordamaglia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Salvatore De Rosis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Asia Spinelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Anna Maria Miglietta
- Breast and General Surgery Unit, Regional Hospital Cosenza, 87100 Cosenza, Italy
| | - Bruno Nardo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Breast and General Surgery Unit, Regional Hospital Cosenza, 87100 Cosenza, Italy
| | | | - Ernestina Marianna De Francesco
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
8
|
Lu Y, Shao Y, Xie Y, Qu H, Qi D, Dong Y, Jin Q, Wang L, Wei J, Quan C. CLDN6 inhibits breast cancer cell malignant behavior by suppressing ERK signaling. Cell Signal 2022; 97:110393. [PMID: 35752352 DOI: 10.1016/j.cellsig.2022.110393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 06/15/2022] [Accepted: 06/19/2022] [Indexed: 11/03/2022]
Abstract
Claudin 6 (CLDN6) is an important component of tight junctions. Through the PDZ binding motif, CLDN6 binds to a variety of signaling proteins that contain the PDZ domain to regulate different signaling pathways, and plays an important role in the occurrence and development of tumors. Our previous work showed that CLDN6 was expressed at low levels in breast cancer cells, and overexpression of CLDN6 inhibited breast cancer cell proliferation, migration and invasion. However, the mechanism of how CLDN6 works remains unclear. In this study, we aimed to explore the mechanism by which CLDN6 inhibits breast cancer cell malignant behavior. As a result, overexpression of CLDN6 inhibited the proliferation of breast cancer cells along with the downregulation of cyclin D1, which plays an important role in regulating cell proliferation. After overexpression of Sp1 in CLDN6-overexpressing cells, the expression of cyclin D1 was upregulated. On the other hand, CLDN6 inhibited breast cancer cell migration and invasion along with the downregulation of IL-8, CXCR2 and FAK. When treated with IL-8, the migration and invasion ability were promoted along with the upregulation of CXCR2 and p-FAK, and the cytoskeleton was rearranged in CLDN6-overexpressing cells. Furthermore, when treated with the ERK signaling activator PMA, the proliferation, migration and invasion abilities were promoted along with the upregulation of Sp1, cyclin D1 and IL-8 in CLDN6-overexpressin cells. In conclusion, CLDN6 suppressed ERK/Sp1/cyclin D1 and ERK/IL-8 signaling to inhibit proliferation, migration and invasion in breast cancer cells. The mechanism may provide experimental evidence for the treatment of breast cancer targeting CLDN6.
Collapse
Affiliation(s)
- Yan Lu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, People's Republic of China
| | - Yijia Shao
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, People's Republic of China
| | - Yinping Xie
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, People's Republic of China
| | - Huinan Qu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, People's Republic of China
| | - Da Qi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, People's Republic of China
| | - Yuan Dong
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, People's Republic of China
| | - Qiu Jin
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, People's Republic of China
| | - Liping Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, People's Republic of China
| | - Junyuan Wei
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, People's Republic of China
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, People's Republic of China.
| |
Collapse
|
9
|
Hozhabri H, Moghaddam MM, Moghaddam MM, Mohammadian A. A comprehensive bioinformatics analysis to identify potential prognostic biomarkers among CC and CXC chemokines in breast cancer. Sci Rep 2022; 12:10374. [PMID: 35725915 PMCID: PMC9209453 DOI: 10.1038/s41598-022-14610-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/06/2022] [Indexed: 11/09/2022] Open
Abstract
Breast cancer (BC) is a major human health problem due to its increasing incidence and mortality rate. CC and CXC chemokines are associated with tumorigenesis and the progression of many cancers. Since the prognostic values of CC and CXC families' expression in various types of cancers are becoming increasingly evident, we aimed to conduct a comprehensive bioinformatics analysis elucidating the prognostic values of the CC and CXC families in BC. Therefore, TCGA, UALCAN, Kaplan–Meier plotter, bc-GenExMiner, cBioPortal, STRING, Enrichr, and TIMER were utilized for analysis. We found that high levels of CCL4/5/14/19/21/22 were associated with better OS and RFS, while elevated expression of CCL24 was correlated with shorter OS in BC patients. Also, high levels of CXCL9/13 indicated longer OS, and enhanced expression of CXCL12/14 was linked with better OS and RFS in BC patients. Meanwhile, increased transcription levels of CXCL8 were associated with worse OS and RFS in BC patients. In addition, our results showed that CCL5, CCL8, CCL14, CCL20, CCL27, CXCL4, and CXCL14 were notably correlated with the clinical outcomes of BC patients. Our findings provide a new point of view that may help the clinical application of CC and CXC chemokines as prognostic biomarkers in BC.
Collapse
Affiliation(s)
- Hossein Hozhabri
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | | | - Madiheh Mazaheri Moghaddam
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Mohammadian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
10
|
Bridoux A, Mousa SA. Screening of a Library for Factor VIIa Inhibitors. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180818666211207125903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
As an alternative to the anticoagulant’s strategy using direct or indirect anti-Xa
drugs, considering other targets upstream in the coagulation cascade such as anti-Factor VIIa could represent
an effective and safer strategy in coagulation and pathological angiogenesis.
Objective:
The objective of the study was to assess a high technology methodology composed of virtual
screening, anticoagulant, and anti-angiogenesis assays to identify potent small-molecule FVIIa inhibitors.
Methods:
Chemical databanks were screened to select molecules bearing functional groups that could fit
into the active site of FVIIa, which were then tested. Ligands assigned with the lowest scores were retained
and then biologically assessed.
Results:
From the 500 molecules considered, 8 chemical structures revealed to be effective compounds in
vitro and to inhibit angiogenesis in the chick chorioallantoic membrane (CAM) model.
Conclusion:
New potent small-molecule FVIIa inhibitors have been identified; further biochemical and
chemical developments would be investigated directly from the selected scaffolds.
Collapse
Affiliation(s)
- Alexandre Bridoux
- Vascular Vision Pharmaceuticals, 5 University Place, Rensselaer, NY 12144, USA
- Pharmaceutical Research Institute,
One Discovery Drive, Rensselaer, NY 12144, USA
| | - Shaker A. Mousa
- Vascular Vision Pharmaceuticals, 5 University Place, Rensselaer, NY 12144, USA
- Pharmaceutical Research Institute,
One Discovery Drive, Rensselaer, NY 12144, USA
| |
Collapse
|
11
|
Mishra A, Suman KH, Nair N, Majeed J, Tripathi V. An updated review on the role of the CXCL8-CXCR1/2 axis in the progression and metastasis of breast cancer. Mol Biol Rep 2021; 48:6551-6561. [PMID: 34426905 DOI: 10.1007/s11033-021-06648-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/11/2021] [Indexed: 12/18/2022]
Abstract
Chronic inflammation is a major factor in tumor growth and progression. Cancer cells secrete C-X-C chemokine ligand 8 (CXCL8) along with its receptor C-X-C chemokine receptor 1 (CXCR1) and chemokine receptor 2 (CXCR2). It plays a significant role in the activation and trafficking of inflammatory mediators, tumor proliferation and interferes in breast cancer development by controlling cell adhesion, proliferation, migration, and metastasis. This axis also plays a significant role in driving different cancers and melanomas, including breast cancer progression, by controlling stem cell masses. Few small-molecule CXCR1/2 inhibitors and CXCL8 releasing inhibitors have been identified in the past two decades that bind these receptors in their inactive forms and blocks their signaling as well as the biological activities associated with inflammation. Inhibitors of certain inflammatory molecules are projected to be more efficient in different inflammatory diseases. Preclinical trials indicate that patients may be benefitted from combined treatment with targeted drugs, chemotherapies, and immunotherapies. Thus, targeting the CXCL8-CXCR1/2 signaling axis in breast cancer could be a promising approach for its therapeutics. This review examines the roles of the CXCL8-CXCR1/2 signaling axis and how it is implicated in the tumor microenvironment in breast cancer. In addition, we also discuss the potential role of the CXCL8-CXCR1/2 axis in targeted therapeutics for breast cancer.
Collapse
Affiliation(s)
- Amaresh Mishra
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201310, India
| | - Kamrul Hassan Suman
- Department of Environment & Aquatic Biology, ABEx Bio-research Center, Azampur, Dhaka, 1230, Bangladesh
| | - Nisha Nair
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences and Research University, Govt of NCT of Delhi, New Delhi, 110017, India
| | - Jaseela Majeed
- School of Allied Health Sciences, Delhi Pharmaceutical Sciences and Research University, Govt of NCT of Delhi, New Delhi, 110017, India
| | - Vishwas Tripathi
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201310, India.
| |
Collapse
|
12
|
Deng F, Weng Y, Li X, Wang T, Fan M, Shi Q. Overexpression of IL-8 promotes cell migration via PI3K-Akt signaling pathway and EMT in triple-negative breast cancer. Pathol Res Pract 2021; 223:152824. [PMID: 34000674 DOI: 10.1016/j.prp.2020.152824] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/18/2019] [Accepted: 01/09/2020] [Indexed: 11/20/2022]
Abstract
Triple-negative breast cancer (TNBC) is a type of malignant and heterogeneous tumor in premenopausal females with ineffective therapeutic targets. IL-8 is one of the earliest discovered chemotaxis cytokines which expression is closely related to the progress of various cancers. Previous studies show that IL-8 determines the prognosis of TNBC patients, nevertheless how IL-8 influence the progress of TNBC is unclear. In our studies, we discovered that overexpression of IL-8 promotes TNBC cells (TNBCs) migration and tumor growth via the PI3K-Akt and MAPK signaling pathway. Cell-cycle of TNBCs arrest at S phase by overexpression of IL-8, however, there is no significant difference on the cell viability and cell apoptosis of TNBCs. Besides, overexpression of IL-8 result in the downregulation of E-cadherin and the upregulation of Cyclin B1 in MDA-MB-231 cells. Taken together, our results suggest that IL-8 plays a crucial role in the progress of TNBC, and it could be a novel therapeutic target of TNBC.
Collapse
Affiliation(s)
- Fang Deng
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, PR China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Yaguang Weng
- Department of Laboratory Medicine, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong, Chongqing 400016, PR China
| | - Xian Li
- Department of Pathology, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong, Chongqing 400016, PR China
| | - Teng Wang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, PR China
| | - Mengtian Fan
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, PR China
| | - Qiong Shi
- Department of Laboratory Medicine, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong, Chongqing 400016, PR China.
| |
Collapse
|
13
|
Mahalakshmi R, Boaz K, Srikant N, Baliga M, Shetty P, Prasad M, Yellapurkar S, Lewis AJ. Neutrophil-to-lymphocyte Ratio: A Surrogate Marker for Prognosis of Oral Squamous Cell Carcinoma. Indian J Med Paediatr Oncol 2021. [DOI: 10.4103/ijmpo.ijmpo_2_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Abstract
Context: Recent studies show that enzymatic contents of the neutrophil granules have a remarkable ability to modulate the tumor microenvironment by causing apoptosis of T-lymphocytes which leaves the host's cell-mediated immunity at stake. The preoperative neutrophil to lymphocyte ratio (NLR) is considered to be an indicator of the immune status of the patients with oral squamous cell carcinoma (OSCC), which will thereby help in predicting the course of the disease. Aims: The aim is to assess the NLR and histopathological prognostic factors pertinent to infiltration of the surrounding structures and correlate them with the clinical prognostic outcomes of OSCC. Settings and Design: This retrospective study involved the retrieval of formalin-fixed, paraffin-embedded, hematoxylin, and eosin-stained sections of 55 cases of OSCC from the departmental archives from 2006 to 2014. Subjects and Methods: Grading of each case was done by Bryne's grading system. The preoperative complete blood counts, relevant case history, and clinical data of the patients involved in the study were collected from the institutional medical records. The NLR was calculated by dividing the serum neutrophil count by the serum lymphocyte count. Statistical Analysis Used: The median NLR was compared between the controls and OSCC cases using Wilcoxon-signed rank test, and the Kaplan-Meier survival analysis was carried out to predict the survival and recurrence status of OSCC. Results: Higher NLR was seen in lymph node and margin-involved cases and also in patients who had tumor recurrence. Kaplan-Meier survival analysis showed that the mean survival dropped from 26 to 4.5 months when NLR ≥5 (P = 0.052). Conclusion: Neutrophil-to-lymphocyte ratio can be used as a prognosticator of survival, recurrence, lymph node status, and margin status in OSCC.
Collapse
Affiliation(s)
- R Mahalakshmi
- Department of Oral Pathology and Microbiology, Manipal College of Dental Sciences, Mangalore, Manipal University, Karnataka, India
| | - Karen Boaz
- Department of Oral Pathology and Microbiology, Manipal College of Dental Sciences, Mangalore, Manipal University, Karnataka, India
| | - N Srikant
- Department of Oral Pathology and Microbiology, Manipal College of Dental Sciences, Mangalore, Manipal University, Karnataka, India
| | - Mohan Baliga
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Mangalore, Manipal University, Karnataka, India
| | - Premalatha Shetty
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Mangalore, Manipal University, Karnataka, India
| | - Mukul Prasad
- Department of Oral Pathology and Microbiology, Manipal College of Dental Sciences, Mangalore, Manipal University, Karnataka, India
| | - Shweta Yellapurkar
- Department of Oral Pathology and Microbiology, Manipal College of Dental Sciences, Mangalore, Manipal University, Karnataka, India
| | - Amitha J Lewis
- Department of Oral Pathology and Microbiology, Manipal College of Dental Sciences, Mangalore, Manipal University, Karnataka, India
| |
Collapse
|
14
|
Haider MT, Ridlmaier N, Smit DJ, Taipaleenmäki H. Interleukins as Mediators of the Tumor Cell-Bone Cell Crosstalk during the Initiation of Breast Cancer Bone Metastasis. Int J Mol Sci 2021; 22:2898. [PMID: 33809315 PMCID: PMC7999500 DOI: 10.3390/ijms22062898] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Patients with advanced breast cancer are at high risk of developing bone metastasis. Despite treatment advances for primary breast cancer, metastatic bone disease remains incurable with a low relative survival. Hence, new therapeutic approaches are required to improve survival and treatment outcome for these patients. Bone is among the most frequent sites of metastasis in breast cancer. Once in the bone, disseminated tumor cells can acquire a dormant state and remain quiescent until they resume growth, resulting in overt metastasis. At this stage the disease is characterized by excessive, osteoclast-mediated osteolysis. Cells of the bone microenvironment including osteoclasts, osteoblasts and endothelial cells contribute to the initiation and progression of breast cancer bone metastasis. Direct cell-to-cell contact as well as soluble factors regulate the crosstalk between disseminated breast cancer cells and bone cells. In this complex signaling network interleukins (ILs) have been identified as key regulators since both, cancer cells and bone cells secrete ILs and express corresponding receptors. ILs regulate differentiation and function of bone cells, with several ILs being reported to act pro-osteoclastogenic. Consistently, the expression level of ILs (e.g., in serum) has been associated with poor prognosis in breast cancer. In this review we discuss the role of the most extensively investigated ILs during the establishment of breast cancer bone metastasis and highlight their potential as therapeutic targets in preventing metastatic outgrowth in bone.
Collapse
Affiliation(s)
- Marie-Therese Haider
- Molecular Skeletal Biology Laboratory, Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.-T.H.); (N.R.)
| | - Nicole Ridlmaier
- Molecular Skeletal Biology Laboratory, Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.-T.H.); (N.R.)
- Department of Life Sciences, IMC FH Krems University of Applied Sciences, 3500 Krems, Austria
| | - Daniel J. Smit
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Hanna Taipaleenmäki
- Molecular Skeletal Biology Laboratory, Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.-T.H.); (N.R.)
| |
Collapse
|
15
|
Wang SF, Huang KH, Tseng WC, Lo JF, Li AFY, Fang WL, Chen CF, Yeh TS, Chang YL, Chou YC, Hung HH, Lee HC. DNAJA3/Tid1 Is Required for Mitochondrial DNA Maintenance and Regulates Migration and Invasion of Human Gastric Cancer Cells. Cancers (Basel) 2020; 12:cancers12113463. [PMID: 33233689 PMCID: PMC7699785 DOI: 10.3390/cancers12113463] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gastric cancer is a common health issue. Deregulated cellular energetics is regarded as a cancer hallmark and mitochondrial dysfunction might contribute to cancer progression. Tid1, a mitochondrial co-chaperone, may play a role as a tumor suppressor in various cancers, but the role of Tid1 in gastric cancers remains under investigated. METHODS The clinical TCGA online database and immunohistochemical staining for Tid1 expression in tumor samples of gastric cancer patients were analyzed. Tid1 knockdown by siRNA was applied to investigate the role of Tid1 in gastric cancer cells. RESULTS Low Tid1 protein-expressing gastric cancer patients had a poorer prognosis and higher lymph node invasion than high Tid1-expressing patients. Knockdown of Tid1 did not increase cell proliferation, colony/tumor sphere formation, or chemotherapy resistance in gastric cancer cells. However, Tid1 knockdown increased cell migration and invasion. Moreover, Tid1 knockdown reduced the mtDNA copy number of gastric cancer cells. In addition, the Tid1-galectin-7-MMP-9 axis might be associated with Tid1 knockdown-induced cell migration and invasion of gastric cancer cells. CONCLUSIONS Tid1 is required for mtDNA maintenance and regulates migration and invasion of gastric cancer cells. Tid1 deletion may be a poor prognostic factor in gastric cancers and could be further investigated for development of gastric cancer treatments.
Collapse
Affiliation(s)
- Sheng-Fan Wang
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei 112, Taiwan; (S.-F.W.); (Y.-L.C.); (Y.-C.C.)
- School of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
| | - Kuo-Hung Huang
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (K.-H.H.); (A.F.-Y.L.); (W.-L.F.)
- Department of Surgery, Division of General Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Wei-Chuan Tseng
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
| | - Jeng-Fan Lo
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
- Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei 112, Taiwan
- Institute of Oral Biology, National Yang-Ming University, Taipei 112, Taiwan
- Cancer Progression Research Center, National Yang-Ming University, Taipei 112, Taiwan;
| | - Anna Fen-Yau Li
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (K.-H.H.); (A.F.-Y.L.); (W.-L.F.)
- Department of Pathology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Wen-Liang Fang
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (K.-H.H.); (A.F.-Y.L.); (W.-L.F.)
- Department of Surgery, Division of General Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Chian-Feng Chen
- Cancer Progression Research Center, National Yang-Ming University, Taipei 112, Taiwan;
| | - Tien-Shun Yeh
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan;
| | - Yuh-Lih Chang
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei 112, Taiwan; (S.-F.W.); (Y.-L.C.); (Y.-C.C.)
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
- Faculty of Pharmacy, National Yang-Ming University, Taipei 112, Taiwan
| | - Yueh-Ching Chou
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei 112, Taiwan; (S.-F.W.); (Y.-L.C.); (Y.-C.C.)
- School of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
- Faculty of Pharmacy, National Yang-Ming University, Taipei 112, Taiwan
| | - Hung-Hsu Hung
- School of Medicine, Faculty of Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Medicine, Division of Gastroenterology, Cheng Hsin General Hospital, Taipei 112, Taiwan
- Correspondence: (H.-H.H.); (H.-C.L.); Tel.: +886-2-2826-7327 (H.-C.L.)
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
- Correspondence: (H.-H.H.); (H.-C.L.); Tel.: +886-2-2826-7327 (H.-C.L.)
| |
Collapse
|
16
|
Deng F, Weng Y, Li X, Wang T, Fan M, Shi Q. Overexpression of IL-8 promotes cell migration via PI3K-Akt signaling pathway and EMT in triple-negative breast cancer. Pathol Res Pract 2020; 216:152902. [PMID: 32147274 DOI: 10.1016/j.prp.2020.152902] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/28/2020] [Accepted: 02/26/2020] [Indexed: 12/17/2022]
Abstract
Triple-negative breast cancer (TNBC) is a type of malignant and heterogeneous tumor in premenopausal females with ineffective therapeutic targets. IL-8 is one of the earliest discovered chemotaxis cytokines which expression is closely related to the progress of various cancers. Previous studies showed that IL-8 determines the prognosis of TNBC patients, nevertheless how IL-8 influences the progress of TNBC is unclear. In our studies, we discovered that overexpression of IL-8 promotes TNBC cells (TNBCs) migration and tumor growth via the PI3K-Akt and MAPK signaling pathway. Cell-cycle of TNBCs arrest at S phase by overexpression of IL-8, however, there is no significant variation on the cell viability and cell apoptosis of TNBCs. Besides, overexpression of IL-8 result in the downregulation of E-cadherin and the upregulation of Cyclin B1 in MDA-MB-231 cells. Taken together, our results suggest that IL-8 performs a crucial role in the progress of TNBC, and it could be a novel therapeutic target of TNBC.
Collapse
Affiliation(s)
- Fang Deng
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, PR China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Yaguang Weng
- Department of Laboratory Medicine, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong, Chongqing, 400016, PR China
| | - Xian Li
- Department of Pathology, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong, Chongqing, 400016, PR China
| | - Teng Wang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, PR China
| | - Mengtian Fan
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, PR China
| | - Qiong Shi
- Department of Laboratory Medicine, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong, Chongqing, 400016, PR China.
| |
Collapse
|
17
|
Aspirin inhibits platelets from reprogramming breast tumor cells and promoting metastasis. Blood Adv 2020; 3:198-211. [PMID: 30670536 DOI: 10.1182/bloodadvances.2018026161] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/16/2018] [Indexed: 12/21/2022] Open
Abstract
It is now recognized that compounds released from tumor cells can activate platelets, causing the release of platelet-derived factors into the tumor microenvironment. Several of these factors have been shown to directly promote neovascularization and metastasis, yet how the feedback between platelet releasate and the tumor cell affects metastatic phenotype remains largely unstudied. Here, we identify that breast tumor cells secrete high levels of interleukin 8 (IL-8, CXCL8) in response to platelet releasate, which promotes their invasive capacity. Furthermore, we found that platelets activate the Akt pathway in breast tumor cells, and inhibition of this pathway eliminated IL-8 production. We therefore hypothesized inhibiting platelets with aspirin could reverse the prometastatic effects of platelets on tumor cell signaling. Platelets treated with aspirin did not activate the Akt pathway, resulting in reduced IL-8 secretion and impaired tumor cell invasion. Of note, patients with breast cancer receiving aspirin had lower circulating IL-8, and their platelets did not increase tumor cell invasion compared with patients not receiving aspirin. Our data suggest platelets support breast tumor metastasis by inducing tumor cells to secrete IL-8. Our data further support that aspirin acts as an anticancer agent by disrupting the communication between platelets and breast tumor cells.
Collapse
|
18
|
He F, Springer NL, Whitman MA, Pathi SP, Lee Y, Mohanan S, Marcott S, Chiou AE, Blank BS, Iyengar N, Morris PG, Jochelson M, Hudis CA, Shah P, Kunitake JAMR, Estroff LA, Lammerding J, Fischbach C. Hydroxyapatite mineral enhances malignant potential in a tissue-engineered model of ductal carcinoma in situ (DCIS). Biomaterials 2019; 224:119489. [PMID: 31546097 PMCID: PMC6878891 DOI: 10.1016/j.biomaterials.2019.119489] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 09/06/2019] [Accepted: 09/11/2019] [Indexed: 01/21/2023]
Abstract
While ductal carcinoma in situ (DCIS) is known as a precursor lesion to most invasive breast carcinomas, the mechanisms underlying this transition remain enigmatic. DCIS is typically diagnosed by the mammographic detection of microcalcifications (MC). MCs consisting of non-stoichiometric hydroxyapatite (HA) mineral are frequently associated with malignant disease, yet it is unclear whether HA can actively promote malignancy. To investigate this outstanding question, we compared phenotypic outcomes of breast cancer cells cultured in control or HA-containing poly(lactide-co-glycolide) (PLG) scaffolds. Exposure to HA mineral in scaffolds increased the expression of pro-tumorigenic interleukin-8 (IL-8) among transformed but not benign cells. Notably, MCF10DCIS.com cells cultured in HA scaffolds adopted morphological changes associated with increased invasiveness and exhibited increased motility that were dependent on IL-8 signaling. Moreover, MCF10DCIS.com xenografts in HA scaffolds displayed evidence of enhanced malignant progression relative to xenografts in control scaffolds. These experimental findings were supported by a pathological analysis of clinical DCIS specimens, which correlated the presence of MCs with increased IL-8 staining and ductal proliferation. Collectively, our work suggests that HA mineral may stimulate malignancy in preinvasive DCIS cells and validate PLG scaffolds as useful tools to study cell-mineral interactions.
Collapse
Affiliation(s)
- Frank He
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Nora L Springer
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA; Department of Diagnostic Medicine/Pathobiology, Kansas State University College of Veterinary Medicine, Manhattan, KS, 66506, USA
| | - Matthew A Whitman
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Siddharth P Pathi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Yeonkyung Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Sunish Mohanan
- Department of Biomedical Sciences, Baker Institute for Animal Health, Cornell University, Ithaca, NY, 14853, USA
| | - Stephen Marcott
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Aaron E Chiou
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Bryant S Blank
- Cornell Center for Animal Resources and Education, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Neil Iyengar
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center/Evelyn H. Lauder Breast and Imaging Center, New York, NY, 10065, USA
| | - Patrick G Morris
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center/Evelyn H. Lauder Breast and Imaging Center, New York, NY, 10065, USA
| | - Maxine Jochelson
- Department of Radiology, Memorial Sloan Kettering Cancer Center/Evelyn H. Lauder Breast and Imaging Center, New York, NY, 10065, USA
| | - Clifford A Hudis
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center/Evelyn H. Lauder Breast and Imaging Center, New York, NY, 10065, USA
| | - Pragya Shah
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA; Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Jennie A M R Kunitake
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14853, USA
| | - Jan Lammerding
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA; Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
19
|
Guo F, Long L, Wang J, Wang Y, Liu Y, Wang L, Luo F. Insights on CXC chemokine receptor 2 in breast cancer: An emerging target for oncotherapy. Oncol Lett 2019; 18:5699-5708. [PMID: 31788042 PMCID: PMC6865047 DOI: 10.3892/ol.2019.10957] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most common malignant neoplasm in women worldwide, and the treatment regimens currently available are far from optimal. Targeted therapy, based on molecular typing of breast cancer, is the most precise form of treatment, and CXC chemokine receptor 2 (CXCR2) is one of the molecular markers used in targeted therapies. As a member of the seven transmembrane G-protein-coupled receptor family, CXCR2 and its associated ligands have been increasingly implicated in tumor-associated processes. These processes include proliferation, angiogenesis, invasion, metastasis, chemoresistance, and stemness and phenotypic maintenance of cancer stem cells. Thus, the inhibition of CXCR2 or its downstream signaling pathways could significantly attenuate tumor progression. Therefore, studies on the biological functions of CXCR2 and its association with neoplasia may help improve the prognosis of breast cancer. Furthermore, the targeting of CXCR2 could supplement the present clinical approaches of breast cancer treatment strategies. The present review discusses the structures and mechanisms of CXCR2 and its ligands. Additionally, the contribution of CXCR2 to the development of breast cancer and its potential therapeutic benefits are also discussed.
Collapse
Affiliation(s)
- Fengzhu Guo
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lang Long
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jiantao Wang
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuyi Wang
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yanyang Liu
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Wang
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Feng Luo
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
20
|
Liubomirski Y, Lerrer S, Meshel T, Rubinstein-Achiasaf L, Morein D, Wiemann S, Körner C, Ben-Baruch A. Tumor-Stroma-Inflammation Networks Promote Pro-metastatic Chemokines and Aggressiveness Characteristics in Triple-Negative Breast Cancer. Front Immunol 2019; 10:757. [PMID: 31031757 PMCID: PMC6473166 DOI: 10.3389/fimmu.2019.00757] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 03/21/2019] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) plays key roles in promoting disease progression in the aggressive triple-negative subtype of breast cancer (TNBC; Basal/Basal-like). Here, we took an integrative approach and determined the impact of tumor-stroma-inflammation networks on pro-metastatic phenotypes in TNBC. With the TCGA dataset we found that the pro-inflammatory cytokines tumor necrosis factor α (TNFα) and interleukin 1β (IL-1β), as well as their target pro-metastatic chemokines CXCL8 (IL-8), CCL2 (MCP-1), and CCL5 (RANTES) were expressed at significantly higher levels in basal patients than luminal-A patients. Then, we found that TNFα- or IL-1β-stimulated co-cultures of TNBC cells (MDA-MB-231, MDA-MB-468, BT-549) with mesenchymal stem cells (MSCs) expressed significantly higher levels of CXCL8 compared to non-stimulated co-cultures or each cell type alone, with or without cytokine stimulation. CXCL8 was also up-regulated in TNBC co-cultures with breast cancer-associated fibroblasts (CAFs) derived from patients. CCL2 and CCL5 also reached the highest expression levels in TNFα/IL-1β-stimulated TNBC:MSC/CAF co-cultures. The elevations in CXCL8 and CCL2 expression partly depended on direct physical contacts between the tumor cells and the MSCs/CAFs, whereas CCL5 up-regulation was entirely dependent on cell-to-cell contacts. Supernatants of TNFα-stimulated TNBC:MSC "Contact" co-cultures induced robust endothelial cell migration and sprouting. TNBC cells co-cultured with MSCs and TNFα gained migration-related morphology and potent migratory properties; they also became more invasive when co-cultured with MSCs/CAFs in the presence of TNFα. Using siRNA to CXCL8, we found that CXCL8 was significantly involved in mediating the pro-metastatic activities gained by TNFα-stimulated TNBC:MSC "Contact" co-cultures: angiogenesis, migration-related morphology of the tumor cells, as well as cancer cell migration and invasion. Importantly, TNFα stimulation of TNBC:MSC "Contact" co-cultures in vitro has increased the aggressiveness of the tumor cells in vivo, leading to higher incidence of mice with lung metastases than non-stimulated TNBC:MSC co-cultures. Similar tumor-stromal-inflammation networks established in-culture with luminal-A cells demonstrated less effective or differently-active pro-metastatic functions than those of TNBC cells. Overall, our studies identify novel tumor-stroma-inflammation networks that may promote TNBC aggressiveness by increasing the pro-malignancy potential of the TME and of the tumor cells themselves, and reveal key roles for CXCL8 in mediating these metastasis-promoting activities.
Collapse
Affiliation(s)
- Yulia Liubomirski
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shalom Lerrer
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tsipi Meshel
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Linor Rubinstein-Achiasaf
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Dina Morein
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Cindy Körner
- Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Adit Ben-Baruch
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
21
|
Interleukin-8 Activates Breast Cancer-Associated Adipocytes and Promotes Their Angiogenesis- and Tumorigenesis-Promoting Effects. Mol Cell Biol 2019; 39:MCB.00332-18. [PMID: 30397072 DOI: 10.1128/mcb.00332-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/25/2018] [Indexed: 01/23/2023] Open
Abstract
Increasing evidence supports the critical role of active stromal adipocytes in breast cancer development and spread. However, the mediators and the mechanisms of action are still elusive. We show here that cancer-associated adipocytes (CAAs) isolated from 10 invasive breast carcinomas are proinflammatory and exhibit active phenotypes, including higher proliferative, invasive, and migratory capacities compared to their adjacent tumor-counterpart adipocytes (TCAs). Furthermore, all CAAs secreted higher level of interleukin-8 (IL-8), which is critical in mediating the paracrine procarcinogenic effects of these cells. Importantly, ectopic expression of IL-8 in TCA cells activated them and enhanced their procarcinogenic effects both in vitro, in a STAT3-dependent manner, and in vivo In contrast, inhibition of the IL-8 signaling using specific short hairpin RNA, anti-IL-8 antibody, or reparixin suppressed the active features of CAAs, including their non-cell-autonomous tumor-promoting activities both on breast luminal cells and in orthotopic tumor xenografts in mice. IL-8 played also an important role in enhancing the proangiogenic effects of breast adipocytes. These results provide clear indication that IL-8 plays key roles in the activation of breast CAAs and acts as a major mediator for their paracrine protumorigenic effects. Thus, targeting CAAs by inhibiting the IL-8 pathway could have great therapeutic value.
Collapse
|
22
|
Upregulated expression of HOXB7 in intrahepatic cholangiocarcinoma is associated with tumor cell metastasis and poor prognosis. J Transl Med 2019; 99:736-748. [PMID: 30664713 PMCID: PMC6760572 DOI: 10.1038/s41374-018-0150-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 02/08/2023] Open
Abstract
Homeobox B7 (HOXB7) protein is reported to be aberrantly expressed in a variety of cancers and to play an important role in multiple cellular processes. However, the specific mechanism by which HOXB7 promotes the malignant progression of intrahepatic cholangiocarcinoma (ICC) remains unclear. Therefore, we used quantitative real-time polymerase chain reaction (PCR) to detect the expression level of HOXB7 in 38 paired ICC tissue samples. Additionally, to assess correlation between HOXB7 and ICC prognosis, we performed immunohistochemistry (IHC) using 122 ICC tissues to detect HOXB7 expression. Cell Counting Kit-8 (CCK-8) and colony formation assays were employed to assess ICC cell proliferation, and Transwell assays were performed to estimate the invasion and migration abilities of ICC cells. The capillary tube formation assay was applied to explore the angiogenic effects of HOXB7. A xenograft tumor model was established in nude mice to assess the role of HOXB7 in tumor growth and lung metastasis. The results showed higher expression of HOXB7 in ICC tissues than in noncancerous tissues, and this increased expression was significantly associated with a poor prognosis. In addition, HOXB7 overexpression enhanced capillary tube formation, invasion and migration of ICC cells in vitro, whereas HOXB7 knockdown produced the opposite results in vitro. Moreover, the role of HOXB7 in promoting tumor growth and metastasis was verified in vivo. Further investigation revealed that the expression levels of MMP2, MMP9, VEGFa, and IL8 were elevated by HOXB7 and that the ERK pathway was activated. Our results demonstrate the prognostic value of HOXB7 and its role in metastasis and angiogenesis in ICC. HOXB7 upregulated MMP2, MMP9, VEGFa, and IL8 expression via the ERK pathway to accelerate the malignant progression of ICC.
Collapse
|
23
|
Abstract
A tumor consists of a group of cells with abnormal growth, capable of acquiring unique characteristics that provide them with the ability to display mercurial migration patterns, adapting to microenvironments and their chemical and physical factors. Interleukins are small proteins secreted mainly by CD3+ and CD4+ T lymphocytes that mediate the "essential for cancer progression" interactions between cells. Interleukins are implicated in both the development and differentiation of different cells (NK, B, and T leukocytes) and, in general, play a major role in many diseases, including breast cancer, due to their unique participation in systemic inflammation and immune system modulation. During the past decade, interleukins proved to be decisive for future immunotherapy, predisposing a more reliable treatment with fewer side effects on normal proliferating cells. The aim of this review is to provide an overview of the role of interleukins implicated in breast cancer progression.
Collapse
Affiliation(s)
| | - George Kolios
- Pharmacology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, GRC
| | | | - Emmanuel N Kontomanolis
- Obstetrics and Gynecology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, GRC
| |
Collapse
|
24
|
Park J, Jang JH, Park GS, Chung Y, You HJ, Kim JH. BLT2, a leukotriene B4 receptor 2, as a novel prognostic biomarker of triple-negative breast cancer. BMB Rep 2018; 51:373-377. [PMID: 29898809 PMCID: PMC6130834 DOI: 10.5483/bmbrep.2018.51.8.127] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is considered to be a notorious type of cancer due to its aggressive metastatic potential and poor prognosis. Recent evidence suggests that BLT2, a low-affinity LTB4 receptor is critically associated with the phenotypes of TNBC cells, including invasion, metastasis, and survival. Furthermore, in a group of 545 breast cancer patients with metastasis, we observed that the high-BLT2 subgroup had a lower disease-free-survival rate than the low-BLT2 subgroup. Thus, we theorized that anti-BLT2 strategies could facilitate the development of new therapies used for TNBC. This review focuses on recent discoveries regarding BLT2 and its roles in as a novel prognostic biomarker in TNBC.
Collapse
Affiliation(s)
- JaeIn Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Jae-Hyun Jang
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Geun-Soo Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Yunro Chung
- Department of Biomedical Informatics, Arizona State University, Scottsdale, AZ 85259, USA
| | - Hye Jin You
- Translational Research Branch, Division of Translational Science, National Cancer Center, Goyang 10408, Korea
| | - Jae-Hong Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| |
Collapse
|
25
|
Wang Y, Tu L, Du C, Xie X, Liu Y, Wang J, Li Z, Jiang M, Cao D, Yan X, Luo F. CXCR2 is a novel cancer stem-like cell marker for triple-negative breast cancer. Onco Targets Ther 2018; 11:5559-5567. [PMID: 30233217 PMCID: PMC6134958 DOI: 10.2147/ott.s174329] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background Breast cancer is the leading cause of mortality from cancer in women worldwide, and cancer stem-like cell (CSC) is responsible for failure treatment of breast cancer. It plays an important role in resistant disease and metastasis. CD44/CD24 and ALDH are well-accepted protein markers of breast CSC, and it was reported that distinct subtypes of breast CSC were identified by the 2 markers. It is possible that there are various kinds of breast CSC which could be identified by different markers, and CSC markers utilized at present are not enough to fully understand breast CSC. Finding out more novel CSC markers is necessary. CXCR2 is involved in breast cancer metastasis, treatment resistance, and recurrence and has positive cross-talk with known breast CSC protein markers. It can be concluded that CXCR2 is related to breast CSC, and further study is in need. Results In this study, we assessed expression of CXCR2 with immunohistochemistry in breast cancer tissues from 37 patients and discovered that level of CXCR2 was significantly lower in triple-negative breast cancer (TNBC) compared with non-TNBC. CXCR2 expression decreased in estrogen receptor-negative or HER2-negative breast cancer, but not progesterone receptor-negative counterparts. By immunofluorescence, we observed high coexpression rate of CXCR2 and CSC-related proteins, including NANOG and SOX2. To prove our speculation that CXCR2 was a novel CSC marker for TNBC, we used 4T1 cell, which is a TNBC cell line, to analyze CXCR2-positive subpopulations and observed that CXCR2-positive 4T1 cells showed characteristics of CSC, including resistance to cisplatinum, radiation, and hypoxia, low proportion (around 1%), much more tumor xenografts, tumor spherule formation, and higher levels of CSC-related mRNA compared with CXCR2-negative cells. Conclusion CXCR2 is an acceptable and newly discovered CSC marker for only TNBC.
Collapse
Affiliation(s)
- Yuyi Wang
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China, ;
| | - Li Tu
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China, ;
| | - Chi Du
- Department of Oncology, The Second People's Hospital of Neijiang, Neijiang, Sichuan, People's Republic of China
| | - Xiaoxiao Xie
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China, ;
| | - Yanyang Liu
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China, ;
| | - Jiantao Wang
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China, ;
| | - Zhixi Li
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China, ;
| | - Ming Jiang
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China, ;
| | - Dan Cao
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China, ;
| | - Xi Yan
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China, ;
| | - Feng Luo
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China, ;
| |
Collapse
|
26
|
Vazquez Rodriguez G, Abrahamsson A, Jensen LDE, Dabrosin C. Adipocytes Promote Early Steps of Breast Cancer Cell Dissemination via Interleukin-8. Front Immunol 2018; 9:1767. [PMID: 30105032 PMCID: PMC6077262 DOI: 10.3389/fimmu.2018.01767] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/17/2018] [Indexed: 12/21/2022] Open
Abstract
Fat is a major tissue component in human breast cancer (BC). Whether breast adipocytes (BAd) affect early stages of BC metastasis is yet unknown. BC progression is dependent on angiogenesis and inflammation, and interleukin-8 (IL-8) and vascular endothelial growth factor (VEGF) are key regulators of these events. Here, we show that BAd increased the dissemination of estrogen receptor positive BC cells (BCC) in vivo in the zebrafish model of metastasis, while dissemination of the more aggressive and metastatic BCC such as estrogen receptor negative was unaffected. While anti-VEGF and anti-IL-8 exhibited equal inhibition of angiogenesis at the primary tumor site, anti-IL-8 reduced BCC dissemination whereas anti-VEGF had minor effects on this early metastatic event. Mechanistically, overexpression of cell-adhesion molecules in BCC and neutrophils via IL-8 increased the dissemination of BCC. Importantly, the extracellular in vivo levels of IL-8 were 40-fold higher than those of VEGF in human BC. Our results suggest that IL-8 is a clinical relevant and promising therapeutic target for human BC.
Collapse
Affiliation(s)
- Gabriela Vazquez Rodriguez
- Department of Oncology, Linköping University, Linköping, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Annelie Abrahamsson
- Department of Oncology, Linköping University, Linköping, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Lasse Dahl Ejby Jensen
- Department of Medical and Health Sciences, Division of Cardiovascular Medicine, Linköping University, Linköping, Sweden
| | - Charlotta Dabrosin
- Department of Oncology, Linköping University, Linköping, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
27
|
Nickel A, Blücher C, Kadri OA, Schwagarus N, Müller S, Schaab M, Thiery J, Burkhardt R, Stadler SC. Adipocytes induce distinct gene expression profiles in mammary tumor cells and enhance inflammatory signaling in invasive breast cancer cells. Sci Rep 2018; 8:9482. [PMID: 29930291 PMCID: PMC6013441 DOI: 10.1038/s41598-018-27210-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 05/25/2018] [Indexed: 12/14/2022] Open
Abstract
Obesity is a known risk factor for breast cancer. Since obesity rates are constantly rising worldwide, understanding the molecular details of the interaction between adipose tissue and breast tumors becomes an urgent task. To investigate potential molecular changes in breast cancer cells induced by co-existing adipocytes, we used a co-culture system of different breast cancer cell lines (MCF-7 and T47D: ER+/PR+/HER2- and MDA-MB-231: ER-/PR-/HER2-) and murine 3T3-L1 adipocytes. Here, we report that co-culture with adipocytes revealed distinct changes in global gene expression pattern in the different breast cancer cell lines. Our microarray data revealed that in both ER+ cell lines, top upregulated genes showed significant enrichment for hormone receptor target genes. In triple-negative MDA-MB-231 cells, co-culture with adipocytes led to the induction of pro-inflammatory genes, mainly involving genes of the Nf-κB signaling pathway. Moreover, co-cultured MDA-MB-231 cells showed increased secretion of the pro-inflammatory interleukins IL-6 and IL-8. Using a specific NF-κB inhibitor, these effects were significantly decreased. Finally, migratory capacities were significantly increased in triple-negative breast cancer cells upon co-culture with adipocytes, indicating an enhanced aggressive cell phenotype. Together, our studies illustrate that factors secreted by adipocytes have a significant impact on the molecular biology of breast cancer cells.
Collapse
Affiliation(s)
- Annina Nickel
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Christina Blücher
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Omaeir Al Kadri
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Nancy Schwagarus
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Silvana Müller
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Michael Schaab
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Joachim Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Ralph Burkhardt
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Sonja C Stadler
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany.
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
28
|
Xu M, Jiang H, Wang H, Liu J, Liu B, Guo Z. SB225002 inhibits prostate cancer invasion and attenuates the expression of BSP, OPN and MMP‑2. Oncol Rep 2018; 40:726-736. [PMID: 29917166 PMCID: PMC6072299 DOI: 10.3892/or.2018.6504] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/31/2018] [Indexed: 11/14/2022] Open
Abstract
The mechanisms of malignant cell metastasis to secondary sites are complex and multifactorial. Studies have demonstrated that small integrin-binding ligand N-linked glycoproteins (SIBLINGs), particularly bone sialoprotein (BSP) and osteopontin (OPN), are involved in neoplastic growth and metastasis. SIBLINGs promote malignant cell invasion and metastasis by enhancing matrix metalloproteinase 2 (MMP-2) and MMP-9 expression. Moreover, BSP and OPN can combine with integrin, which is located on the tumor cell surface, to further promote the malignant behavior of tumor cells. In the present study, we investigated whether SB225002, a specific CXCR2 receptor antagonist, can inhibit prostate cancer cell expression of BSP and OPN and reduce cancer cell invasion ability. A series of experiments showed that after SB225002 treatment, the proliferation, invasion and migration of two androgen-independent prostate cancer cell lines were inhibited, but this inhibitory effect was not observed on androgen-dependent prostate cancer cells. Western blotting showed that the PI3K signaling pathway could regulate the expression of SIBLING and MMP family proteins, and SB22055 could reduce the expression of BSP, OPN and MMP-2 in prostate cancer cells by inhibiting AKT/mTOR phosphorylation. Finally, in vivo experiments confirmed that SB225002 inhibited the proliferation of prostate cancer cells in vivo, and the expression levels of BSP, OPN and MMP-2 were also inhibited.
Collapse
Affiliation(s)
- Meng Xu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Huamao Jiang
- Graduate School of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Haiguang Wang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Jiajie Liu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Baohao Liu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Zhongqiang Guo
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
29
|
Ji S, Zhang W, Zhang X, Hao C, Hao A, Gao Q, Zhang H, Sun J, Hao J. Sohlh2 suppresses epithelial to mesenchymal transition in breast cancer via downregulation of IL-8. Oncotarget 2018; 7:49411-49424. [PMID: 27384482 PMCID: PMC5226517 DOI: 10.18632/oncotarget.10355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 06/16/2016] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is one of the deadliest cancers worldwide due to its strong metastasis to other organs. Metastasis of breast cancer involves a complex set of events, including epithelial-mesenchymal transition (EMT) that increases invasiveness of the tumor cells. We previously identified sohlh2 is a tumor suppressor in the pathogenesis of ovarian cancer. However, the functions of sohlh2 in breast cancer cell migration and invasion remain unknown. Here we report a novel sohlh2/IL-8 signaling pathway in the invasive breast cancer. We observed sohlh2 expression was downregulated in the metastatic breast cancer. Ectopic sohlh2 expression in breast cancer cells reduced EMT and inhibited cell migration and invasion in vitro, and metastasis in vivo. Moreover, the depletion of sohlh2 induced the opposite effects to ectopic sohlh2 expression. RNA-Seq data from a sohlh2 knockdown breast cancer cell line showed that after sohlh2 depletion, the mRNA level of interleukin 8 (IL-8) was significantly increased in these cancer cells, which consequently increased secretion of IL-8 protein. Using chromatin immunoprecipitation and reporter assays, we demonstrated that sohlh2 bound to IL-8 promoter and repressed its activities. The enhanced migration and invasion in sohlh2 -ablated MCF-7 cells were blocked by knockdown of IL-8 expression, while exogenous IL-8 neutralized the anti-migratory and invasive activities of sohlh2 in MDA-MB-231cells. Overall, these results demonstrate that sohlh2 functions as a tumor metastasis suppressor via suppressing IL-8 expression in breast cancer.
Collapse
Affiliation(s)
- Shufang Ji
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan 250012, PR China
| | - Wenfang Zhang
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan 250012, PR China
| | - Xiaoli Zhang
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan 250012, PR China
| | - Chunyan Hao
- Department of Pathology, School of Medicine, Shandong University, Jinan 250012, PR China
| | - Aijun Hao
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan 250012, PR China
| | - Qing Gao
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan 250012, PR China
| | - Hongying Zhang
- Department of Biology, Jinan Vocational College of Nursing, Jinan 250000, PR China
| | - Jinhao Sun
- Department of Human Anatomy, School of Medicine, Shandong University, Jinan 250012, PR China
| | - Jing Hao
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan 250012, PR China
| |
Collapse
|
30
|
Tobouti PL, Bolt R, Radhakrishnan R, de Sousa SCOM, Hunter KD. Altered Toll-like receptor expression and function in HPV-associated oropharyngeal carcinoma. Oncotarget 2018; 9:236-248. [PMID: 29416610 PMCID: PMC5787461 DOI: 10.18632/oncotarget.18959] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 06/16/2017] [Indexed: 12/25/2022] Open
Abstract
Toll-like receptors (TLRs) have been widely investigated due to their importance in the inflammatory response and possible links to tumor promotion/regression and prognosis. In cancers with an infective etiology, such as human papillomavirus (HPV)-associated Oropharyngeal Squamous Cell Carcinoma (OPSCC), TLR responses may be activated and play a role in tumorigenesis. Our aim was to assess the expression of all TLRs in OPSCC cell lines (both HPV+ and HPV-) by qPCR, Western Blot and flow cytometry and assess their response to TLR ligands lipopolysaccharide (LPS), LPS ultra-pure (LPS-UP) and peptidoglycan (PGN) by analyzing IL-8 and IL-6 production. We also immunostained 61 OPSCC tissue samples with anti-TLR4. Results showed lower TLR1 and TLR6 mRNA expression and higher TLR9 protein expression in HPV+ when compared to HPV-OPSCC cells. TLR4 expression did not vary by HPV status in OPSCC cells, but TLR4 expression was significantly lower in HPV+OPSCC tissues. After stimulation with PGN, only one cell line (HPV+) did not secrete IL-6 or IL-8. Furthermore, HPV+OPSCC lines showed no IL-6 or IL-8 production on treatment with LPS/LPS-UP. The data suggest changes in TLR4 signaling in HPV+OPSCC, since we have shown lower tissue expression of TLR4 and no pro-inflammatory response after stimulation with LPS and LPS-UP. Also, it suggests that OPSCC may respond to HPV infection by increased expression of TLR9. This study demonstrates differences in expression and function of TLRs in OPSCC, which are dependent on HPV status, and may indicate subversion of the innate immune response by HPV infection.
Collapse
Affiliation(s)
- Priscila Lie Tobouti
- Oral Pathology Department, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Robert Bolt
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Raghu Radhakrishnan
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal University, Manipal, India
| | | | - Keith D. Hunter
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| |
Collapse
|
31
|
Zhao Z, Wang S, Lin Y, Miao Y, Zeng Y, Nie Y, Guo P, Jiang G, Wu J. Epithelial-mesenchymal transition in cancer: Role of the IL-8/IL-8R axis. Oncol Lett 2017; 13:4577-4584. [PMID: 28599458 DOI: 10.3892/ol.2017.6034] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 01/19/2017] [Indexed: 12/26/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a biological process that is associated with cancer metastasis and invasion. In cancer, EMT promotes cell motility, invasion and distant metastasis. Interleukin (IL)-8 is highly expressed in tumors and may induce EMT. The IL-8/IL-8R axis has a vital role in EMT in carcinoma, which is regulated by several signaling pathways, including the transforming growth factor β-spleen associated tyrosine kinase/Src-AKT/extracellular signal-regulated kinase, p38/Jun N-terminal kinase-activating transcription factor-2, phosphoinositide 3-kinase/AKT, nuclear factor-κB and Wnt signaling pathways. Blocking the IL-8/IL-8R signaling pathway may be a novel strategy to reduce metastasis and improve patient survival rates. This review will cover IL-8-IL-8R signaling pathway in tumor epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Zhiwei Zhao
- West China Medical Center, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shichao Wang
- West China Medical Center, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,School of Basic Medicine, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Yingbo Lin
- Department of Oncology and Pathology, Karolinska Institute, Cancer Centre Karolinska, SE-171 76 Stockholm, Sweden
| | - Yali Miao
- West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ye Zeng
- West China Medical Center, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yongmei Nie
- School of Basic Medicine, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Peng Guo
- West China Medical Center, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Guangyao Jiang
- Outpatient Building, West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jiang Wu
- West China Medical Center, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,School of Basic Medicine, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| |
Collapse
|
32
|
Wu F, Chen W, Gillis B, Fischbach C, Estroff LA, Gourdon D. Protein-crystal interface mediates cell adhesion and proangiogenic secretion. Biomaterials 2017; 116:174-185. [PMID: 27940370 PMCID: PMC5223748 DOI: 10.1016/j.biomaterials.2016.11.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/30/2016] [Accepted: 11/24/2016] [Indexed: 11/26/2022]
Abstract
The nanoscale materials properties of bone apatite crystals have been implicated in breast cancer bone metastasis and their interactions with extracellular matrix proteins are likely involved. In this study, we used geologic hydroxyapatite (HAP, Ca10(PO4)6(OH)2), closely related to bone apatite, to investigate how HAP surface chemistry and nano/microscale topography individually influence the crystal-protein interface, and how the altered protein deposition impacts subsequent breast cancer cell activities. We first utilized Förster resonance energy transfer (FRET) to assess the molecular conformation of fibronectin (Fn), a major extracellular matrix protein upregulated in cancer, when it adsorbed onto HAP facets. Our analysis reveals that both low surface charge density and nanoscale roughness of HAP facets individually contributed to molecular unfolding of Fn. We next quantified cell adhesion and secretion on Fn-coated HAP facets using MDA-MB-231 breast cancer cells. Our data show elevated proangiogenic and proinflammatory secretions associated with more unfolded Fn adsorbed onto nano-rough HAP facets with low surface charge density. These findings not only deconvolute the roles of crystal surface chemistry and topography in interfacial protein deposition but also enhance our knowledge of protein-mediated breast cancer cell interactions with apatite, which may be implicated in tumor growth and bone metastasis.
Collapse
Affiliation(s)
- Fei Wu
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA.
| | - Weisi Chen
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Brian Gillis
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Ithaca, NY 14853, USA
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Ithaca, NY 14853, USA
| | - Delphine Gourdon
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA; Department of Physics, University of Ottawa, Ottawa, ON K1N 6N5, Canada.
| |
Collapse
|
33
|
Lesage J, Suarez‐Carmona M, Neyrinck‐Leglantier D, Grelet S, Blacher S, Hunziker W, Birembaut P, Noël A, Nawrocki‐Raby B, Gilles C, Polette M. Zonula occludens‐1/NF‐κB/CXCL8: a new regulatory axis for tumor angiogenesis. FASEB J 2017; 31:1678-1688. [DOI: 10.1096/fj.201600890r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/03/2017] [Indexed: 01/18/2023]
Affiliation(s)
- Julien Lesage
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
| | - Meggy Suarez‐Carmona
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Deborah Neyrinck‐Leglantier
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
| | - Simon Grelet
- Department of Biochemistry and Molecular BiologyMedical University of South Carolina Charleston South Carolina USA
| | - Silvia Blacher
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Walter Hunziker
- Epithelial Cell Biology LaboratoryInstitute of Molecular and Cell Biology Singapore Singapore
| | - Philippe Birembaut
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
- Laboratory of BiopathologyCentres Hospitaliers Universitaires Reims France
| | - Agnes Noël
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Béatrice Nawrocki‐Raby
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
| | - Christine Gilles
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Myriam Polette
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
- Laboratory of BiopathologyCentres Hospitaliers Universitaires Reims France
| |
Collapse
|
34
|
Wei JD, Jang JH, Kim JH. RanBPM inhibits BLT2-mediated IL-8 production and invasiveness in aggressive breast cancer cells. Biochem Biophys Res Commun 2016; 483:305-311. [PMID: 28027932 DOI: 10.1016/j.bbrc.2016.12.147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022]
Abstract
RanBPM is a scaffolding protein that regulates several cellular processes by interacting with various proteins. Previously, we reported that RanBPM acts as a negative regulator of BLT2, a low-affinity leukotriene B4 receptor; thus, it interferes with BLT2-mediated cell motility. In the present study, we observed that the expression levels of RanBPM were markedly reduced in the highly aggressive MDA-MB-435 and MDA-MB-231 human breast cancer cell lines compared with those in non-invasive MCF-7 cells. Additionally, we found that the restoration of RanBPM levels suppressed the invasiveness of these aggressive breast cancer cells in a manner dependent on BLT2 activation. In contrast, the knockdown of endogenous RanBPM by shRNA strongly promoted invasiveness in non-invasive MCF-7 cells. We also observed that RanBPM suppressed the invasiveness of aggressive breast cancer cells by inhibiting BLT2-mediated reactive oxygen species (ROS) generation and IL-8 production. Taken together, our results suggest that RanBPM acts as a negative regulator of BLT2, thus attenuating the invasiveness of aggressive breast cancer cells.
Collapse
Affiliation(s)
- Jun-Dong Wei
- School of Life Sciences and Biotechnology, Korea University, 5-1 Anam-dong, Sungbuk-gu, Seoul, 02841, South Korea
| | - Jae-Hyun Jang
- School of Life Sciences and Biotechnology, Korea University, 5-1 Anam-dong, Sungbuk-gu, Seoul, 02841, South Korea
| | - Jae-Hong Kim
- School of Life Sciences and Biotechnology, Korea University, 5-1 Anam-dong, Sungbuk-gu, Seoul, 02841, South Korea.
| |
Collapse
|
35
|
Nakayama J, Ito E, Fujimoto J, Watanabe S, Semba K. Comparative analysis of gene regulatory networks of highly metastatic breast cancer cells established by orthotopic transplantation and intra-circulation injection. Int J Oncol 2016; 50:497-504. [PMID: 28000849 DOI: 10.3892/ijo.2016.3809] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 10/21/2016] [Indexed: 11/06/2022] Open
Abstract
Metastasis signature genes in breast cancer have been studied comparing transcriptomic profiles of highly metastatic cancer cell lines established by intra-circulation injection with that of their parental cell line. However, this method is not suitable to analyze the initial steps of metastasis including invasion into local tissues and the circulatory system. To characterize the molecular mechanisms of early metastasis, we established highly metastatic MDA-MB-231 cell lines that metastasized to lung by the two animal transplantation models: the orthotopic transplantation method, which mimics all steps of metastasis, or intra-circulation injection method. We then performed data-mining and network analysis of gene expression profiles of metastatic cell lines established by each transplantation method. Transcriptome analysis of seven metastatic cell lines revealed novel lung metastasis signature genes, including known metastasis promoting genes and signature genes. In the OXconc (orthotopic xenograft concentration) signature, 'chemotaxis' and 'cell adhesion' terms were enriched. In the TVIconc (tail vein injection concentration) signature, 'antigen recognition' and 'cell adhesion' were enriched. Furthermore, network analysis of the metastasis signature genes highlighted hub genes in the gene regulatory network. Our findings show that expression profiles of highly metastatic cell lines were different between the orthotopic transplantation and intra-circulation injection method. It also indicates that some metastatic signature genes have been missed in previous studies. Characterization of metastasis genes using the orthotopic transplantation method will be helpful in understanding the multi-step mechanisms of metastasis. Signature genes in OXconc may have the potential to become prognostic markers.
Collapse
Affiliation(s)
- Jun Nakayama
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Emi Ito
- Division of Informatics, Translational Research Center (Tokyo Branch), Fukushima Medical University, Tokyo, Japan
| | - Jiro Fujimoto
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Shinya Watanabe
- Division of Informatics, Translational Research Center (Tokyo Branch), Fukushima Medical University, Tokyo, Japan
| | - Kentaro Semba
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| |
Collapse
|
36
|
Selitrennik M, Lev S. PYK2 integrates growth factor and cytokine receptors signaling and potentiates breast cancer invasion via a positive feedback loop. Oncotarget 2016; 6:22214-26. [PMID: 26084289 PMCID: PMC4673158 DOI: 10.18632/oncotarget.4257] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/29/2015] [Indexed: 12/27/2022] Open
Abstract
The involvement of ErbB family members in breast cancer progression and metastasis has been demonstrated by many studies. However, the downstream effectors that mediate their migratory and invasive responses have not been fully explored. In this study, we show that the non-receptor tyrosine kinase PYK2 is a key effector of EGFR and HER2 signaling in human breast carcinoma. We found that PYK2 is activated by both EGF and heregulin (HRG) in breast cancer cells, and positively regulates EGF/HRG-induced cell spreading, migration and invasion. PYK2 depletion markedly affects ERK1/2 and STAT3 phosphorylation in response to EGF/HRG as well as to IL8 treatment. Importantly, PYK2 depletion also reduced EGF/HRG-induced MMP9 and IL8 transcription, while IL8 inhibition abrogated EGF-induced MMP9 transcription and attenuated cell invasion. IL8, which is transcriptionally regulated by STAT3 and induces PYK2 activation, prolonged EGF-induced PYK2, STAT3 and ERK1/2 phosphorylation suggesting that IL8 acts through an autocrine loop to reinforce EGF-induced signals. Collectively our studies suggest that PYK2 is a common downstream effector of ErbB and IL8 receptors, and that PYK2 integrates their signaling pathways through a positive feedback loop to potentiate breast cancer invasion. Hence, PYK2 could be a potential therapeutic target for a subset of breast cancer patients.
Collapse
Affiliation(s)
- Michael Selitrennik
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Sima Lev
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
37
|
Liu Q, Li A, Tian Y, Wu JD, Liu Y, Li T, Chen Y, Han X, Wu K. The CXCL8-CXCR1/2 pathways in cancer. Cytokine Growth Factor Rev 2016; 31:61-71. [PMID: 27578214 PMCID: PMC6142815 DOI: 10.1016/j.cytogfr.2016.08.002] [Citation(s) in RCA: 436] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/13/2016] [Accepted: 08/23/2016] [Indexed: 02/07/2023]
Abstract
Persistent infection or chronic inflammation contributes significantly to tumourigenesis and tumour progression. C-X-C motif ligand 8 (CXCL8) is a chemokine that acts as an important multifunctional cytokine to modulate tumour proliferation, invasion and migration in an autocrine or paracrine manner. Studies have suggested that CXCL8 and its cognate receptors, C-X-C chemokine receptor 1 (CXCR1) and CX-C chemokine receptor 2 (CXCR2), mediate the initiation and development of various cancers including breast cancer, prostate cancer, lung cancer, colorectal carcinoma and melanoma. CXCL8 also integrates with multiple intracellular signalling pathways to produce coordinated effects. Neovascularisation, which provides a basis for fostering tumour growth and metastasis, is now recognised as a critical function of CXCL8 in the tumour microenvironment. In this review, we summarize the biological functions and ficlinical significance of the CXCL8 signalling axis in cancer. We also propose that CXCL8 may be a potential therapeutic target for cancer treatment
Collapse
Affiliation(s)
- Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Anping Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yijun Tian
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jennifer D Wu
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Yu Liu
- Department of Geriatric, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tengfei Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yuan Chen
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
38
|
Kolb R, Sutterwala FS, Zhang W. Obesity and cancer: inflammation bridges the two. Curr Opin Pharmacol 2016; 29:77-89. [PMID: 27429211 DOI: 10.1016/j.coph.2016.07.005] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/05/2016] [Accepted: 07/06/2016] [Indexed: 12/14/2022]
Abstract
Obesity is a growing public health problem and affects 35% US adults. Obesity increases the risk of many cancer types and is associated with poor outcomes. Clinical management of cancer patients has been essentially the same between normal weight and obese individuals. Understanding causal mechanisms by which obesity drives cancer initiation and progression is essential for the development of novel precision therapy for obese cancer patients. One caveat is that various mechanisms have been proposed for different cancer types for their progression under obesity. Since obesity is known to have global impact on inflammation, here we will summarize recent literature and discuss the potential of inflammation being the common causal mechanism to promote cancer promotion across cancer types.
Collapse
Affiliation(s)
- Ryan Kolb
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Fayyaz S Sutterwala
- Division of Infectious Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Weizhou Zhang
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Medical Scientist Training Program, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Free Radical and Radiation Biology Program, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Cancer Genes and Pathway Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
39
|
Peroxiredoxin 1 interacts with and blocks the redox factor APE1 from activating interleukin-8 expression. Sci Rep 2016; 6:29389. [PMID: 27388124 PMCID: PMC4937415 DOI: 10.1038/srep29389] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 06/20/2016] [Indexed: 01/18/2023] Open
Abstract
APE1 is an essential DNA repair protein that also possesses the ability to regulate transcription. It has a unique cysteine residue C65, which maintains the reduce state of several transcriptional activators such as NF-κB. How APE1 is being recruited to execute the various biological functions remains unknown. Herein, we show that APE1 interacts with a novel partner PRDX1, a peroxidase that can also prevent oxidative damage to proteins by serving as a chaperone. PRDX1 knockdown did not interfere with APE1 expression level or its DNA repair activities. However, PRDX1 knockdown greatly facilitates APE1 detection within the nucleus by indirect immunofluorescence analysis, even though APE1 level was unchanged. The loss of APE1 interaction with PRDX1 promotes APE1 redox function to activate binding of the transcription factor NF-κB onto the promoter of a target gene, the proinflammatory chemokine IL-8 involved in cancer invasion and metastasis, resulting in its upregulation. Depletion of APE1 blocked the upregulation of IL-8 in the PRDX1 knockdown cells. Our findings suggest that the interaction of PRDX1 with APE1 represents a novel anti-inflammatory function of PRDX1, whereby the association safeguards APE1 from reducing transcription factors and activating superfluous gene expression, which otherwise could trigger cancer invasion and metastasis.
Collapse
|
40
|
Sharma B, Varney ML, Saxena S, Wu L, Singh RK. Induction of CXCR2 ligands, stem cell-like phenotype, and metastasis in chemotherapy-resistant breast cancer cells. Cancer Lett 2016; 372:192-200. [PMID: 26797460 PMCID: PMC4821546 DOI: 10.1016/j.canlet.2015.12.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/04/2015] [Accepted: 12/07/2015] [Indexed: 12/30/2022]
Abstract
CXCR2 and its ligands have been shown to play an important role in tumor angiogenesis, therapy resistance and progression. In this study, we investigated whether CXCR2 ligands are responsible for the survival advantage and metastasis of drug-resistant cells and examined the underlying mechanism(s) doxorubicin or paclitaxel resistant mammary tumor cells. Our results demonstrated that drug-resistant Cl66 cells upregulated CXCR2 ligands but downregulated expression of CXCR2. We observed delayed tumor growth but increased metastasis in mice using these drug-resistant cells. Furthermore, we observed differential upregulation of stem cell and mesenchymal markers in the doxorubicin and paclitaxel-resistant tumor cells. Abrogation of the CXCR2 signaling axis using CXCR2 ligand neutralization resulted in significant inhibition of drug-resistant cell growth. Together, our data suggest chemotherapy-specific differential regulation of CXCR2 ligands, stem cell-like and mesenchymal phenotypes, and enhanced metastasis in drug-resistant cells and targeting CXCR2 signaling, may help circumvent therapy resistance in breast cancer.
Collapse
Affiliation(s)
- Bhawna Sharma
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE; Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Michelle L Varney
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Sugandha Saxena
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Lingyun Wu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Rakesh K Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE; Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE.
| |
Collapse
|
41
|
Brenner DR, Brockton NT, Kotsopoulos J, Cotterchio M, Boucher BA, Courneya KS, Knight JA, Olivotto IA, Quan ML, Friedenreich CM. Breast cancer survival among young women: a review of the role of modifiable lifestyle factors. Cancer Causes Control 2016; 27:459-72. [PMID: 26970739 PMCID: PMC4796361 DOI: 10.1007/s10552-016-0726-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 02/06/2016] [Indexed: 02/06/2023]
Abstract
Almost 7 % of breast cancers are diagnosed among women age 40 years and younger in Western populations. Clinical outcomes among young women are worse. Early age-of-onset increases the risk of contralateral breast cancer, local and distant recurrence, and subsequent mortality. Breast cancers in young women (BCYW) are more likely to present with triple-negative (TNBC), TP53-positive, and HER-2 over-expressing tumors than among older women. However, despite these known differences in breast cancer outcomes and tumor subtypes, there is limited understanding of the basic biology, epidemiology, and optimal therapeutic strategies for BCYW. Several modifiable lifestyle factors associated with reduced risk of developing breast cancer have also been implicated in improved prognosis among breast cancer survivors of all ages. Given the treatment-related toxicities and the extended window for late effects, long-term lifestyle modifications potentially offer significant benefits to BCYW. In this review, we propose a model identifying three main areas of lifestyle factors (energy imbalance, inflammation, and dietary nutrient adequacy) that may influence survival in BCYW. In addition, we provide a summary of mechanisms of action and a synthesis of previous research on each of these topics.
Collapse
Affiliation(s)
- Darren R Brenner
- Department of Cancer Epidemiology and Prevention Research, CancerControl Alberta, Alberta Health Services, Room 513, Holy Cross Centre, Box ACB, 2210-2nd St. SW, Calgary, AB, T2S 3C3, Canada. .,Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada. .,Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Nigel T Brockton
- Department of Cancer Epidemiology and Prevention Research, CancerControl Alberta, Alberta Health Services, Room 513, Holy Cross Centre, Box ACB, 2210-2nd St. SW, Calgary, AB, T2S 3C3, Canada.,Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Joanne Kotsopoulos
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.,Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Michelle Cotterchio
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.,Prevention and Cancer Control, Cancer Care Ontario, Toronto, ON, Canada
| | - Beatrice A Boucher
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada.,Prevention and Cancer Control, Cancer Care Ontario, Toronto, ON, Canada
| | - Kerry S Courneya
- Faculty of Physical Education and Recreation, University of Alberta, Edmonton, AB, Canada
| | - Julia A Knight
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Ivo A Olivotto
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - May Lynn Quan
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Christine M Friedenreich
- Department of Cancer Epidemiology and Prevention Research, CancerControl Alberta, Alberta Health Services, Room 513, Holy Cross Centre, Box ACB, 2210-2nd St. SW, Calgary, AB, T2S 3C3, Canada.,Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
42
|
Lau WH, Pandey V, Kong X, Wang XN, Wu Z, Zhu T, Lobie PE. Trefoil Factor-3 (TFF3) Stimulates De Novo Angiogenesis in Mammary Carcinoma both Directly and Indirectly via IL-8/CXCR2. PLoS One 2015; 10:e0141947. [PMID: 26559818 PMCID: PMC4641663 DOI: 10.1371/journal.pone.0141947] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/13/2015] [Indexed: 12/19/2022] Open
Abstract
Mammary carcinoma cells produce pro-angiogenic factors to stimulate angiogenesis and tumor growth. Trefoil factor-3 (TFF3) is an oncogene secreted from mammary carcinoma cells and associated with poor prognosis. Herein, we demonstrate that TFF3 produced in mammary carcinoma cells functions as a promoter of tumor angiogenesis. Forced expression of TFF3 in mammary carcinoma cells promoted proliferation, survival, invasion and in vitro tubule formation of human umbilical vein endothelial cells (HUVEC). MCF7-TFF3 cells with forced expression of TFF3 generated tumors with enhanced microvessel density as compared to tumors formed by vector control cells. Depletion of TFF3 in mammary carcinoma cells by siRNA concordantly decreased the angiogenic behavior of HUVEC. Forced expression of TFF3 in mammary carcinoma cells stimulated IL-8 transcription and subsequently enhanced IL-8 expression in both mammary carcinoma cells and HUVEC. Depletion of IL-8 in mammary carcinoma cells with forced expression of TFF3, or antibody inhibition of IL-8, partially abrogated mammary carcinoma cell TFF3-stimulated HUVEC angiogenic behavior in vitro, as did inhibition of the IL-8 receptor, CXCR2. Depletion of STAT3 by siRNA in MCF-7 cells with forced expression of TFF3 partially diminished the angiogenic capability of TFF3 on stimulation of cellular processes of HUVEC. Exogenous recombinant hTFF3 also directly promoted the angiogenic behavior of HUVEC. Hence, TFF3 is a potent angiogenic factor and functions as a promoter of de novo angiogenesis in mammary carcinoma, which may co-coordinate with the growth promoting and metastatic actions of TFF3 in mammary carcinoma to enhance tumor progression.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Blotting, Western
- Breast Neoplasms/blood supply
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation/genetics
- Cells, Cultured
- Coculture Techniques
- Female
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Interleukin-8/genetics
- Interleukin-8/metabolism
- MCF-7 Cells
- Mice, Inbred BALB C
- Mice, Nude
- Microscopy, Fluorescence
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Peptides/genetics
- Peptides/metabolism
- RNA Interference
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Transplantation, Heterologous
- Trefoil Factor-3
Collapse
Affiliation(s)
- Wai-Hoe Lau
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Vijay Pandey
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Xiangjun Kong
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Xiao-Nan Wang
- Department of Pathology, Anhui Medical University, Hefei, Anhui, PR China
- Laboratory of Pathogenic Microbiology and Immunology, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - ZhengSheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui, PR China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
- * E-mail: (PEL); (TZ)
| | - Peter E Lobie
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- National University Cancer Institute of Singapore, National Health System, National University of Singapore, Singapore, Singapore
- * E-mail: (PEL); (TZ)
| |
Collapse
|
43
|
Choi S, Coonrod S, Estroff L, Fischbach C. Chemical and physical properties of carbonated hydroxyapatite affect breast cancer cell behavior. Acta Biomater 2015; 24:333-42. [PMID: 26072364 DOI: 10.1016/j.actbio.2015.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 05/06/2015] [Accepted: 06/01/2015] [Indexed: 01/04/2023]
Abstract
Breast microcalcifications are routinely explored for mammographic detection of breast cancer and are primarily composed of non-stoichiometric hydroxyapatite (Ca10-x(PO4)6-x(CO3)x(OH)2-x) (HA). Interestingly, HA morphology and carbonate substitution vary in malignant vs. benign lesions. However, whether or not these changes (i) are functionally linked and (ii) impact malignancy remains unclear due in part to lack of model systems that permit evaluating these possibilities. Here, we have adapted a 96 well-based mineralized culture platform to investigate breast cancer cell behavior in response to systematic changes in the chemical and physical properties of HA. By adjusting the carbonate content of the simulated body fluid (SBF) solutions used during growth, we can control the morphology and carbonate substitution of the deposited HA. Our results suggest that both the combined and individual effects of these differences alter breast cancer cell growth and secretion of tumorigenic interleukin-8 (IL-8). Consequently, changes in both HA carbonate incorporation and morphology impact the behavior of breast cancer cells. Collectively, our data underline the importance of biomineralized culture platforms to evaluate the functional contribution of HA material properties to the pathogenesis of breast cancer. STATEMENT OF SIGNIFICANCE Breast microcalcifications are small mineral deposits primarily composed of hydroxyapatite (HA). HA physicochemical properties have been of considerable interest, as these are often altered during breast cancer progression and linked to malignancy. However, the functional relationship between these changes and malignancy remains unclear due in part to lack of model systems. Here, we have adapted a previously developed a 96 well-based culture platform to evaluate breast cancer cell behavior in response to systematic changes in HA properties. Our results demonstrate that changes in HA morphology and carbonate content influence breast cancer cell growth and interleukin-8 secretion, and suggest that characterizing the effect of HA properties on breast cancer cells may improve our understanding of breast cancer development and progression.
Collapse
|
44
|
Di Maggio FM, Minafra L, Forte GI, Cammarata FP, Lio D, Messa C, Gilardi MC, Bravatà V. Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond) 2015; 12:14. [PMID: 25705130 PMCID: PMC4336767 DOI: 10.1186/s12950-015-0058-3] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 01/29/2015] [Indexed: 01/05/2023] Open
Abstract
Ionizing radiation (IR) activates both pro-and anti-proliferative signal pathways producing an imbalance in cell fate decision. IR is able to regulate several genes and factors involved in cell-cycle progression, survival and/or cell death, DNA repair and inflammation modulating an intracellular radiation-dependent response. Radiation therapy can modulate anti-tumour immune responses, modifying tumour and its microenvironment. In this review, we report how IR could stimulate inflammatory factors to affect cell fate via multiple pathways, describing their roles on gene expression regulation, fibrosis and invasive processes. Understanding the complex relationship between IR, inflammation and immune responses in cancer, opens up new avenues for radiation research and therapy in order to optimize and personalize radiation therapy treatment for each patient.
Collapse
Affiliation(s)
- Federica Maria Di Maggio
- />Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | - Luigi Minafra
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | - Giusi Irma Forte
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | | | - Domenico Lio
- />Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
| | - Cristina Messa
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
- />Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy
- />Nuclear Medicine Center, San Gerardo Hospital, Monza, Italy
| | - Maria Carla Gilardi
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
- />Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy
- />Nuclear Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Bravatà
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| |
Collapse
|
45
|
Li H, Bergeron S, Annis MG, Siegel PM, Juncker D. Serial analysis of 38 proteins during the progression of human breast tumor in mice using an antibody colocalization microarray. Mol Cell Proteomics 2015; 14:1024-37. [PMID: 25680959 PMCID: PMC4390249 DOI: 10.1074/mcp.m114.046516] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Indexed: 01/20/2023] Open
Abstract
Proteins in serum or plasma hold great potential for use in disease diagnosis and monitoring. However, the correlation between tumor burden and protein biomarker concentration has not been established. Here, using an antibody colocalization microarray, the protein concentration in serum was measured and compared with the size of mammary xenograft tumors in 11 individual mice from the time of injection; seven blood samples were collected from each tumor-bearing mouse as well as control mice on a weekly basis. The profiles of 38 proteins detected in sera from these animals were analyzed by clustering, and we identified 10 proteins with the greatest relative increase in serum concentration that correlated with growth of the primary mammary tumor. To evaluate the diagnosis of cancer based on these proteins using either an absolute threshold (i.e. a concentration cutoff) or self-referenced differential threshold based on the increase in concentration before cell injection, receiver operating characteristic curves were produced for 10 proteins with increased concentration, and the area under curve was calculated for each time point based on a single protein or on a panel of proteins, in each case showing a rapid increase of the area under curve. Next, the sensitivity and specificity of individual and optimal protein panels were calculated, showing high accuracy as early as week 2. These results provide a foundation for studies of tumor growth through measuring serial changes of protein concentration in animal models.
Collapse
Affiliation(s)
- Huiyan Li
- From the ‡Biomedical Engineering Department, §McGill University and Genome Quebec Innovation Centre
| | - Sébastien Bergeron
- From the ‡Biomedical Engineering Department, §McGill University and Genome Quebec Innovation Centre
| | | | - Peter M Siegel
- ‖Rosalind and Morris Goodman Cancer Research Centre, and
| | - David Juncker
- From the ‡Biomedical Engineering Department, §McGill University and Genome Quebec Innovation Centre, **Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec H3A 0G1, Canada
| |
Collapse
|
46
|
Deng Y, Zhang Y, Sun S, Wang Z, Wang M, Yu B, Czajkowsky DM, Liu B, Li Y, Wei W, Shi Q. An integrated microfluidic chip system for single-cell secretion profiling of rare circulating tumor cells. Sci Rep 2014; 4:7499. [PMID: 25511131 PMCID: PMC4266859 DOI: 10.1038/srep07499] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/27/2014] [Indexed: 02/08/2023] Open
Abstract
Genetic and transcriptional profiling, as well as surface marker identification of single circulating tumor cells (CTCs) have been demonstrated. However, quantitatively profiling of functional proteins at single CTC resolution has not yet been achieved, owing to the limited purity of the isolated CTC populations and a lack of single-cell proteomic approaches to handle and analyze rare CTCs. Here, we develop an integrated microfluidic system specifically designed for streamlining isolation, purification and single-cell secretomic profiling of CTCs from whole blood. Key to this platform is the use of photocleavable ssDNA-encoded antibody conjugates to enable a highly purified CTC population with <75 ‘contaminated' blood cells. An enhanced poly-L-lysine barcode pattern is created on the single-cell barcode chip for efficient capture rare CTC cells in microchambers for subsequent secreted protein profiling. This system was extensively evaluated and optimized with EpCAM-positive HCT116 cells seeded into whole blood. Patient blood samples were employed to assess the utility of the system for isolation, purification and single-cell secretion profiling of CTCs. The CTCs present in patient blood samples exhibit highly heterogeneous secretion profile of IL-8 and VEGF. The numbers of secreting CTCs are found not in accordance with CTC enumeration based on immunostaining in the parallel experiments.
Collapse
Affiliation(s)
- Yuliang Deng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yu Zhang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Shuai Sun
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Zhihua Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Minjiao Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Beiqin Yu
- Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Daniel M Czajkowsky
- 1] Center for Bio-Detection and Bio-Instrumentation, Shanghai Jiao Tong University, Shanghai, China [2] School of Biomedicial Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Bingya Liu
- 1] Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China [2] Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Li
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Wei Wei
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Qihui Shi
- 1] Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China [2] State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, China [3] Center for Bio-Detection and Bio-Instrumentation, Shanghai Jiao Tong University, Shanghai, China [4] School of Biomedicial Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
47
|
Singh JK, Simões BM, Howell SJ, Farnie G, Clarke RB. Recent advances reveal IL-8 signaling as a potential key to targeting breast cancer stem cells. Breast Cancer Res 2014; 15:210. [PMID: 24041156 PMCID: PMC3978717 DOI: 10.1186/bcr3436] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Breast cancer stem-like cells (CSCs) are an important therapeutic target as they are purported to be responsible for tumor initiation, maintenance, metastases, and disease recurrence. Interleukin-8 (IL-8) is upregulated in breast cancer compared with normal breast tissue and is associated with poor prognosis. IL-8 is reported to promote breast cancer progression by increasing cell invasion, angiogenesis, and metastases and is upregulated in HER2-positive cancers. Recently, we and others have established that IL-8 via its cognate receptors, CXCR1 and CXCR2, is also involved in regulating breast CSC activity. Our work demonstrates that in metastatic breast CSCs, CXCR1/2 signals via transactivation of HER2. Given the importance of HER2 in breast cancer and in regulating CSC activity, a pathway driving the activation of these receptors would have important biological and clinical consequences, especially in tumors that express high levels of IL-8 and other CXCR1/2-activating ligands. Here, we review the IL-8 signaling pathway and the role of HER2 in maintaining an IL-8 inflammatory loop and discuss the potential of combining CXCR1/2 inhibitors with other treatments such as HER2-targeted therapy as a novel approach to eliminate CSCs and improve patient survival.
Collapse
|
48
|
Yin J, Zeng F, Wu N, Kang K, Yang Z, Yang H. Interleukin-8 promotes human ovarian cancer cell migration by epithelial-mesenchymal transition induction in vitro. Clin Transl Oncol 2014; 17:365-70. [PMID: 25373532 DOI: 10.1007/s12094-014-1240-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 10/01/2014] [Indexed: 12/14/2022]
Abstract
PURPOSE It has been well established that high serum levels of interleukin-8 (IL-8) in ovarian cancer result in a poor clinical outcome. Thus, the aim of this study was investigating the role of IL-8 in ovarian cancer development. METHODS Two human ovarian cancer cell lines (SKOV3 and OVCAR3) were cocultured with IL-8 (100 ng/L) for 24 h, then cell migration was determined by transwell assay. Epithelial-mesenchymal transition (EMT)-associated proteins including E-cadherin and β-catenin, and phosphorylation status of β-catenin were investigated by Western blot analysis. RESULTS After treatment with IL-8 (100 ng/L) for 24 h, transwell assay result showed that the number of migrated ovarian cells increased significantly. Western blot analysis revealed that protein levels of E-cadherin were decreased, while that of β-catenin were elevated both in IL-8 pretreated SKOV3 and OVCAR3 cells. We further found that phosphorylation status of β-catenin were elevated either in cytoplasm or in nucleus of these two ovarian cancer cell lines after treatment with IL-8 for 24 h. CONCLUSIONS Our data suggest that IL-8 induces EMT in ovarian cancer cells and implicates its potential role in enhancing ovarian cancer cell metastasis.
Collapse
Affiliation(s)
- J Yin
- Department of Gynecology, The Ninth People's Hospital of Chongqing, 69 JiaLing Road, Chongqing, 400700, People's Republic of China
| | | | | | | | | | | |
Collapse
|
49
|
Malonia SK, Yadav B, Sinha S, Lazennec G, Chattopadhyay S. Chromatin remodeling protein SMAR1 regulates NF-κB dependent Interleukin-8 transcription in breast cancer. Int J Biochem Cell Biol 2014; 55:220-6. [PMID: 25239884 DOI: 10.1016/j.biocel.2014.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 08/14/2014] [Accepted: 09/08/2014] [Indexed: 11/15/2022]
Abstract
Interleukin-8 (IL-8) is a pleiotropic chemokine involved in metastasis and angiogenesis of breast tumors. The expression of IL-8 is deregulated in metastatic breast carcinomas owing to aberrant NF-κB activity, which is known to positively regulate IL-8 transcription. Earlier, we have shown that tumor suppressor SMAR1 suppresses NF-κB transcriptional activity by modulating IκBα function. Here, we show that NF-κB target gene IL-8, is a direct transcriptional target of SMAR1. Using chromatin immunoprecipitation and reporter assays, we demonstrate that SMAR1 binds to IL-8 promoter MAR (matrix attachment region) and recruits HDAC1 dependent co-repressor complex. Further, we also show that SMAR1 antagonizes p300-mediated acetylation of RelA/p65, a post-translational modification indispensable for IL-8 transactivation. Thus, we decipher a new role of SMAR1 in NF-κB dependent transcriptional regulation of pro-angiogenic chemokine IL-8.
Collapse
Affiliation(s)
- Sunil K Malonia
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Bhawna Yadav
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Surajit Sinha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Gwendel Lazennec
- INSERM, U844, University of Montpellier, Montpellier F-34091, France
| | | |
Collapse
|
50
|
Inflammatory factors of the tumor microenvironment induce plasticity in nontransformed breast epithelial cells: EMT, invasion, and collapse of normally organized breast textures. Neoplasia 2014; 15:1330-46. [PMID: 24403855 DOI: 10.1593/neo.131688] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 10/27/2013] [Accepted: 10/29/2013] [Indexed: 12/12/2022] Open
Abstract
Nontransformed breast epithelial cells that are adjacent to tumor cells are constantly exposed to tumor necrosis factor-α (TNFα) and interleukin-1β (IL-1β), two inflammatory cytokines identified as having pro-tumoral causative roles. We show that continuous stimulation of nontransformed breast epithelial cells by TNFα + IL-1β for 2 to 3 weeks induced their spreading and epithelial-to-mesenchymal transition (EMT). The mechanistic bases for this slow induction of EMT by TNFα + IL-1β are: 1) it took 2 to 3 weeks for the cytokines to induce the expression of the EMT activators Zeb1 and Snail; 2) although Twist has amplified the EMT-inducing activities of Zeb1 + Snail, its expression was reduced by TNFα + IL-1β; however, the lack of Twist was compensated by prolonged stimulation with TNFα + IL-1β that has potentiated the EMT-inducing activities of Zeb1 + Snail. Stimulation by TNFα + IL-1β has induced the following dissemination-related properties in the nontransformed cells: 1) up-regulation of functional matrix metalloproteinases; 2) induction of migratory and invasive capabilities; 3) disruption of the normal phenotype of organized three-dimensional acini structures typically formed only by nontransformed breast cells and spreading of nontransformed cells out of such acini. Our findings suggest that TNFα + IL-1β induce dissemination of nontransformed breast epithelial cells and their reseeding at the primary tumor site; if, then, such detached cells are exposed to transforming events, they may form secondary malignant focus and lead to disease recurrence. Thus, our study reveals novel pathways through which the inflammatory microenvironment may contribute to relapsed disease in breast cancer.
Collapse
|