1
|
D'Addario CA, Matsumura S, Kitagawa A, Lainer GM, Zhang F, D'silva M, Khan MY, Froogh G, Gruzdev A, Zeldin DC, Schwartzman ML, Gupte SA. Global and endothelial G-protein coupled receptor 75 (GPR75) knockout relaxes pulmonary artery and mitigates hypoxia-induced pulmonary hypertension. Vascul Pharmacol 2023; 153:107235. [PMID: 37742819 PMCID: PMC10841449 DOI: 10.1016/j.vph.2023.107235] [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: 10/11/2022] [Revised: 09/04/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
RATIONALE Pulmonary hypertension (PH) is a multifactorial disease with a poor prognosis and inadequate treatment options. We found two-fold higher expression of the orphan G-Protein Coupled Receptor 75 (GPR75) in leukocytes and pulmonary arterial smooth muscle cells from idiopathic PH patients and from lungs of C57BL/6 mice exposed to hypoxia. We therefore postulated that GPR75 signaling is critical to the pathogenesis of PH. METHODS To test this hypothesis, we exposed global (Gpr75-/-) and endothelial cell (EC) GPR75 knockout (EC-Gpr75-/-) mice and wild-type (control) mice to hypoxia (10% oxygen) or normal atmospheric oxygen for 5 weeks. We then recorded echocardiograms and performed right heart catheterizations. RESULTS Chronic hypoxia increased right ventricular systolic and diastolic pressures in wild-type mice but not Gpr75-/- or EC-Gpr75-/- mice. In situ hybridization and qPCR results revealed that Gpr75 expression was increased in the alveoli, airways and pulmonary arteries of mice exposed to hypoxia. In addition, levels of chemokine (CC motif) ligand 5 (CCL5), a low affinity ligand of GPR75, were increased in the lungs of wild-type, but not Gpr75-/-, mice exposed to hypoxia, and CCL5 enhanced hypoxia-induced contraction of intra-lobar pulmonary arteries in a GPR75-dependent manner. Gpr75 knockout also increased pulmonary cAMP levels and decreased contraction of intra-lobar pulmonary arteries evoked by endothelin-1 or U46619 in cAMP-protein kinase A-dependent manner. CONCLUSION These results suggest GPR75 has a significant role in the development of hypoxia-induced PH.
Collapse
Affiliation(s)
| | - Shun Matsumura
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Atsushi Kitagawa
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Gregg M Lainer
- Department of Cardiology, and Heart and Vascular Institute, Westchester Medical Center and New York Medical College, Valhalla, NY 10595, USA
| | - Frank Zhang
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Melinee D'silva
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Mohammad Y Khan
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Ghezal Froogh
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Artiom Gruzdev
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Darryl C Zeldin
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | | | - Sachin A Gupte
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA.
| |
Collapse
|
2
|
Zhang XF, Zhang XL, Wang YJ, Fang Y, Li ML, Liu XY, Luo HY, Tian Y. The regulatory network of the chemokine CCL5 in colorectal cancer. Ann Med 2023; 55:2205168. [PMID: 37141250 PMCID: PMC10161960 DOI: 10.1080/07853890.2023.2205168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
The chemokine CCL5 plays a potential role in the occurrence and development of colorectal cancer (CRC). Previous studies have shown that CCL5 directly acts on tumor cells to change tumor metastatic rates. In addition, CCL5 recruits immune cells and immunosuppressive cells into the tumor microenvironment (TME) and reshapes the TME to adapt to tumor growth or increase antitumor immune efficacy, depending on the type of secretory cells releasing CCL5, the cellular function of CCL5 recruitment, and the underlying mechanisms. However, at present, research on the role played by CCL5 in the occurrence and development of CRC is still limited, and whether CCL5 promotes the occurrence and development of CRC and its role remain controversial. This paper discusses the cells recruited by CCL5 in patients with CRC and the specific mechanism of this recruitment, as well as recent clinical studies of CCL5 in patients with CRC.Key MessagesCCL5 plays dual roles in colorectal cancer progression.CCL5 remodels the tumor microenvironment to adapt to colorectal cancer tumor growth by recruiting immunosuppressive cells or by direct action.CCL5 inhibits colorectal cancer tumor growth by recruiting immune cells or by direct action.
Collapse
Affiliation(s)
- Xin-Feng Zhang
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiao-Li Zhang
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ya-Jing Wang
- Department of General Surgery, Third Medical Center of PLA General Hospital, Beijing, China
| | - Yuan Fang
- Organ Transplant Department, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Meng-Li Li
- Honghui Hospital affiliated to Yunnan University, Kunming, China
| | - Xing-Yu Liu
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hua-You Luo
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yan Tian
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
3
|
Navarro HI, Liu Y, Fraser A, Lefaudeux D, Chia JJ, Vong L, Roifman CM, Hoffmann A. RelB-deficient autoinflammatory pathology presents as interferonopathy, but in mice is interferon-independent. J Allergy Clin Immunol 2023; 152:1261-1272. [PMID: 37460023 PMCID: PMC10858800 DOI: 10.1016/j.jaci.2023.06.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/19/2023] [Accepted: 06/13/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND Autoimmune diseases are leading causes of ill health and morbidity and have diverse etiology. Two signaling pathways are key drivers of autoimmune pathology, interferon and nuclear factor-κB (NF-κB)/RelA, defining the 2 broad labels of interferonopathies and relopathies. Prior work has established that genetic loss of function of the NF-κB subunit RelB leads to autoimmune and inflammatory pathology in mice and humans. OBJECTIVE We sought to characterize RelB-deficient autoimmunity by unbiased profiling of the responses of immune sentinel cells to stimulus and to determine the functional role of dysregulated gene programs in the RelB-deficient pathology. METHODS Transcriptomic profiling was performed on fibroblasts and dendritic cells derived from patients with RelB deficiency and knockout mice, and transcriptomic responses and pathology were assessed in mice deficient in both RelB and the type I interferon receptor. RESULTS We found that loss of RelB in patient-derived fibroblasts and mouse myeloid cells results in elevated induction of hundreds of interferon-stimulated genes. Removing hyperexpression of the interferon-stimulated gene program did not ameliorate the autoimmune pathology of RelB knockout mice. Instead, we found that RelB suppresses a different set of inflammatory response genes in a manner that is independent of interferon signaling but associated with NF-κB binding motifs. CONCLUSION Although transcriptomic profiling would describe RelB-deficient autoimmune disease as an interferonopathy, the genetic evidence indicates that the pathology in mice is interferon-independent.
Collapse
Affiliation(s)
- Héctor I Navarro
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, Calif; Molecular Biology Institute, Los Angeles, Calif
| | - Yi Liu
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, Calif; Molecular Biology Institute, Los Angeles, Calif; DeepKinase Biotechnologies, Ltd, Beijing, China
| | - Anna Fraser
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, Calif; Molecular Biology Institute, Los Angeles, Calif; Institute for Quantitative and Computational Biosciences, Los Angeles, Calif
| | - Diane Lefaudeux
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, Calif; Institute for Quantitative and Computational Biosciences, Los Angeles, Calif
| | - Jennifer J Chia
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, Calif; Molecular Biology Institute, Los Angeles, Calif; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Calif
| | - Linda Vong
- Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Canada
| | - Chaim M Roifman
- Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Canada
| | - Alexander Hoffmann
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, Calif; Molecular Biology Institute, Los Angeles, Calif; Institute for Quantitative and Computational Biosciences, Los Angeles, Calif.
| |
Collapse
|
4
|
Li C, Xu Y, Zhang J, Zhang Y, He W, Ju J, Wu Y, Wang Y. The effect of resveratrol, curcumin and quercetin combination on immuno-suppression of tumor microenvironment for breast tumor-bearing mice. Sci Rep 2023; 13:13278. [PMID: 37587146 PMCID: PMC10432483 DOI: 10.1038/s41598-023-39279-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/22/2023] [Indexed: 08/18/2023] Open
Abstract
Resveratrol, curcumin, and quercetin are the secondary metabolites from medicinal food homology plants, that have been proven their potency in cancer treatment. However, the antitumor effect of a single component is weak. So, herein, we designed an antitumor compound named RCQ composed of resveratrol, curcumin, and quercetin. This study examined the effect on tumorigenesis and development of 4T1 breast cancer-bearing mice following administering RCQ by intragastric administration. RCQ increased the recruitment of T cells and reduced the accumulation of neutrophils and macrophages in the tumor microenvironment. Meanwhile, RCQ suppressed the development of tumor-infiltrating lymphocytes into immunosuppressive cell subpopulations, including CD4+ T cells to T helper Type 2 type (Th2), tumor-associated neutrophils (TANs) to the N2 TANs, and tumor-associated macrophages (TAMs) cells to M2 TAMs. RCQ reversed the predominance of immunosuppressive infiltrating cells in the tumor microenvironment and tipped the immune balance toward an immune activation state. In vitro the study showed that RCQ significantly increased reactive oxygen species (ROS), reduce mitochondrial membrane potentials in cancer cells, and modulate pro-apoptotic Bcl-2 family members. In conclusion, RCQ can promote the ROS apoptosis mechanism of tumor cells and alleviate immunosuppression of the tumor microenvironment to enhance the anti-tumor effect.
Collapse
Affiliation(s)
- Chenchen Li
- School of Medicine and School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, People's Republic of China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, International Associated Research Center for Intelligent Human Computer Collaboration on Tumor Precision Medicine, School of Pharmacy and The First Affiliated Hospital, Hainan Medical University, Haikou, 571199, Hainan, China
| | - Yajun Xu
- School of Medicine and School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Junfeng Zhang
- School of Medicine and School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Yuxi Zhang
- School of Medicine and School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Wen He
- School of Medicine and School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Jiale Ju
- School of Medicine and School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Yinghua Wu
- School of Medicine and School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Yanli Wang
- School of Medicine and School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, People's Republic of China.
| |
Collapse
|
5
|
Huang Y, Wu L, Sun Y, Li J, Mao N, Yang Y, Zhao M, Ren S. CCL5 might be a prognostic biomarker and associated with immuno-therapeutic efficacy in cancers: A pan-cancer analysis. Heliyon 2023; 9:e18215. [PMID: 37519664 PMCID: PMC10375802 DOI: 10.1016/j.heliyon.2023.e18215] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 08/01/2023] Open
Abstract
Purpose Chemokine ligand 5 (CCL5), a vital member of the CC chemokine family, plays diverse roles in tumorigenesis, metastasis, and prognosis in various human tumors. However, no pan-cancer analysis has been conducted to illustrate its distinctive effects on clinical prognosis via underlying mechanisms and biological characteristics. Methods Herein, we exploited the existed public bioinformatics database, primarily TCGA database and GTEx data, to comprehensively analyze the value of CCL5 involved in patient prognosis. Results This study found that CCL5 was excessively expressed in most tumors and significantly associated with clinical prognosis in 10 out of 33 types of tumors. Notably, CCL5 might be an independent predictive biomarker of clinical outcome in SKCM patients, confirmed by univariate and multivariate Cox regression analysis. Furthermore, we acquired the genetic alteration status of CCL5 in multiple types of tumor tissues from TCGA cohorts. We revealed a potential correlation between the expression level of CCL5 and tumor mutational burden in 33 types of tumors. In addition, data showed that DNA methylation was associated with CCL5 gene expression in THCA, PRAD, LUSC, and BRCA cancers. Immune infiltration and immune checkpoints are fine indexes for evaluating immunotherapy. We uncovered that CCL5 was negatively correlated with the immune infiltration of CD8+ T cell, CD4+ T cell, macrophages, and gamma delta T cells in BRCA-basal and CESC tumors, while a significant positive correlation was observed in BLCA, COAD and other 7 types of tumors. Besides, CCL5 was closely associated with the immune checkpoint molecules in 8 types of tumors. The TIDE score was less in the CCL5 high-expressed group than in the CCL5 low-expressed group in SKCM patients, which indicated that CCL5 might be a fine monitor of immune response for immunotherapy. GO enrichment analysis data uncovered that cytokine-cytokine receptor interaction and chemokine signaling might be involved in the role of CCL5 in regulating tumor pathogenesis and prognosis. Conclusion In conclusion, CCL5 was preliminarly identified as a biomarker of immune response and prognosis for tumors patients via our first comprehensive pan-cancer analysis.
Collapse
Affiliation(s)
- Yanchun Huang
- Department of Laboratory Medicine, The First People's Hospital of Longquanyi District, Chengdu, Chengdu 610100, China
- Department of Laboratory Medicine, West China Longquan Hospital Sichuan University, Chengdu 610100, China
| | - Lijuan Wu
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yong Sun
- Department of Laboratory Medicine, The First People's Hospital of Longquanyi District, Chengdu, Chengdu 610100, China
- Department of Laboratory Medicine, West China Longquan Hospital Sichuan University, Chengdu 610100, China
| | - Jiwen Li
- Department of Laboratory Medicine, The First People's Hospital of Longquanyi District, Chengdu, Chengdu 610100, China
- Department of Laboratory Medicine, West China Longquan Hospital Sichuan University, Chengdu 610100, China
| | - Nan Mao
- Department of Nephrology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Yeqing Yang
- Department of Oncology, Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou 646000, China
| | - Ming Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Sichong Ren
- Department of Nephrology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| |
Collapse
|
6
|
Zhang Y, Wang W, Min J, Liu S, Wang Q, Wang Y, Xiao Y, Li X, Zhou Z, Liu S. ZNF451 favors triple-negative breast cancer progression by enhancing SLUG-mediated CCL5 transcriptional expression. Cell Rep 2023; 42:112654. [PMID: 37342906 DOI: 10.1016/j.celrep.2023.112654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 05/01/2023] [Accepted: 06/01/2023] [Indexed: 06/23/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype with limited effective therapies because of the absence of definitive targets. Here, we demonstrate that the expression of ZNF451, a poorly characterized vertebrate zinc-finger protein, is upregulated in TNBC and associated with a poor prognosis. Elevated ZNF451 expression facilitates TNBC progression by interacting with and enhancing the activity of the transcriptional activator snail family transcriptional repressor 2 (SLUG). Mechanistically, the ZNF451-SLUG complex preferentially recruits the acetyltransferase p300/CBP-associated factor (PCAF) to the CCL5 promoter, selectively facilitating CCL5 transcription by enhancing the acetylation of SLUG and local chromatin, leading to recruitment and activation of tumor-associated macrophages (TAMs). Disturbing the ZNF451-SLUG interaction using a peptide suppresses TNBC progression by reducing CCL5 expression and counteracting the migration and activation of TAMs. Collectively, our work provides mechanistic insights into the oncogene-like functions of ZNF451 and suggests that ZNF451 is a potential target for development of effective therapies against TNBC.
Collapse
Affiliation(s)
- Yu Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Wanyu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiali Min
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Suosi Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Qianrong Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yu Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yang Xiao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China.
| |
Collapse
|
7
|
Figy C, Guo A, Fernando VR, Furuta S, Al-Mulla F, Yeung KC. Changes in Expression of Tumor Suppressor Gene RKIP Impact How Cancers Interact with Their Complex Environment. Cancers (Basel) 2023; 15:cancers15030958. [PMID: 36765912 PMCID: PMC9913418 DOI: 10.3390/cancers15030958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/20/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Tumor microenvironment (TME) is the immediate environment where cancer cells reside in a tumor. It is composed of multiple cell types and extracellular matrix. Microenvironments can be restrictive or conducive to the progression of cancer cells. Initially, microenvironments are suppressive in nature. Stepwise accumulation of mutations in oncogenes and tumor suppressor genes enables cancer cells to acquire the ability to reshape the microenvironment to advance their growth and metastasis. Among the many genetic events, the loss-of-function mutations in tumor suppressor genes play a pivotal role. In this review, we will discuss the changes in TME and the ramifications on metastasis upon altered expression of tumor metastasis suppressor gene RKIP in breast cancer cells.
Collapse
Affiliation(s)
- Christopher Figy
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH 43614, USA
| | - Anna Guo
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH 43614, USA
| | - Veani Roshale Fernando
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH 43614, USA
| | - Saori Furuta
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH 43614, USA
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait City 15462, Kuwait
| | - Kam C. Yeung
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH 43614, USA
- Correspondence:
| |
Collapse
|
8
|
Augimeri G, Fiorillo M, Morelli C, Panza S, Giordano C, Barone I, Catalano S, Sisci D, Andò S, Bonofiglio D. The Omega-3 Docosahexaenoyl Ethanolamide Reduces CCL5 Secretion in Triple Negative Breast Cancer Cells Affecting Tumor Progression and Macrophage Recruitment. Cancers (Basel) 2023; 15:cancers15030819. [PMID: 36765778 PMCID: PMC9913844 DOI: 10.3390/cancers15030819] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/03/2023] Open
Abstract
Triple-negative breast cancer (TNBC), an aggressive breast cancer subtype lacking effective targeted therapies, is considered to feature a unique cellular microenvironment with high infiltration of tumor-associated macrophages (TAM), which contribute to worsening breast cancer patient outcomes. Previous studies have shown the antitumoral actions of the dietary omega-3 docosahexaenoic acid (DHA) in both tumor epithelial and stromal components of the breast cancer microenvironment. Particularly in breast cancer cells, DHA can be converted into its conjugate with ethanolamine, DHEA, leading to a more effective anti-oncogenic activity of the parent compound in estrogen receptor-positive breast cancer cells. Here, we investigated the ability of DHEA to attenuate the malignant phenotype of MDA-MB-231 and MDA-MB-436 TNBC cell lines, which in turn influenced TAM behaviors. Our findings revealed that DHEA reduced the viability of TNBC cells in a concentration-dependent manner and compromised cell migration and invasion. Interestingly, DHEA inhibited oxygen consumption and extracellular acidification rates, reducing respiration and the glycolytic reserve in both cell lines. In a co-culture system, TNBC cells exposed to DHEA suppressed recruitment of human THP-1 cells, reduced their viability, and the expression of genes associated with TAM phenotype. Interestingly, we unraveled that the effects of DHEA in TNCB cells were mediated by reduced C-C motif chemokine ligand 5 (CCL5) expression and secretion affecting macrophage recruitment. Overall, our data, shedding new light on the antitumoral effects of DHA ethanolamine-conjugated, address this compound as a promising option in the treatment of TNBC patients.
Collapse
Affiliation(s)
- Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Marco Fiorillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Catia Morelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Salvatore Panza
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Correspondence: (C.G.); (D.B.)
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Diego Sisci
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Correspondence: (C.G.); (D.B.)
| |
Collapse
|
9
|
Takada YK, Fujita M, Takada Y. Pro-Inflammatory Chemokines CCL5, CXCL12, and CX3CL1 Bind to and Activate Platelet Integrin αIIbβ3 in an Allosteric Manner. Cells 2022; 11:cells11193059. [PMID: 36231020 PMCID: PMC9563052 DOI: 10.3390/cells11193059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/16/2022] Open
Abstract
Activation of platelet integrin αIIbβ3, a key event for hemostasis and thrombus formation, is known to be mediated exclusively by inside-out signaling. We showed that inflammatory chemokines CX3CL1 and CXCL12 in previous studies, and CCL5 in this study, bound to the allosteric binding site (site 2) of vascular integrin αvβ3, in addition to the classical ligand binding site (site 1), and allosterically activated integrins independent of inside-out signaling. Since αIIbβ3 is exposed to inflammatory chemokines at increased concentrations during inflammation (e.g., cytokine/chemokine storm) and platelet activation, we hypothesized that these chemokines bind to and activate αIIbβ3 in an allosteric activation mechanism. We found that these chemokines bound to αIIbβ3. Notably, they activated soluble αIIbβ3 in 1 mM Ca2+ by binding to site 2. They activated cell-surface αIIbβ3 on CHO cells, which lack machinery for inside-out signaling or chemokine receptors, quickly (<1 min) and at low concentrations (1–10 ng/mL) compared to activation of soluble αIIbβ3, probably because chemokines bind to cell surface proteoglycans. Furthermore, activation of αIIbβ3 by the chemokines was several times more potent than 1 mM Mn2+. We propose that CCL5 and CXCL12 (stored in platelet granules) may allosterically activate αIIbβ3 upon platelet activation and trigger platelet aggregation. Transmembrane CX3CL1 on activated endothelial cells may mediate platelet–endothelial interaction by binding to and activating αIIbβ3. Additionally, these chemokines in circulation over-produced during inflammation may trigger αIIbβ3 activation, which is a possible missing link between inflammation and thrombosis.
Collapse
Affiliation(s)
- Yoko K. Takada
- Department of Dermatology, School of Medicine, University of California–Davis, 4645 Second Ave., Research III Suite 3300, Sacramento, CA 95817, USA
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Masaaki Fujita
- Department of Dermatology, School of Medicine, University of California–Davis, 4645 Second Ave., Research III Suite 3300, Sacramento, CA 95817, USA
| | - Yoshikazu Takada
- Department of Dermatology, School of Medicine, University of California–Davis, 4645 Second Ave., Research III Suite 3300, Sacramento, CA 95817, USA
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Correspondence:
| |
Collapse
|
10
|
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
|
11
|
Hu A, Zhu J, Zeng C, Lin CH, Yu J, Liu JQ, Lynch K, Talebian F, Pan X, Yan J, Dong Y, Li Z, Bai XF. IL-27 Induces CCL5 Production by T Lymphocytes, Which Contributes to Antitumor Activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2239-2245. [PMID: 35418466 PMCID: PMC9050872 DOI: 10.4049/jimmunol.2100885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/25/2022] [Indexed: 04/15/2023]
Abstract
IL-27 is a pleiotropic cytokine that exhibits stimulatory/regulatory functions on multiple lineages of immune cells including T lymphocytes. In this study, we demonstrate that IL-27 directly induces CCL5 production by T lymphocytes, particularly CD8+ T cells in vitro and in vivo. IL-27-induced CCL5 production is IL-27R-dependent. In CD4+ T cells, IL-27-induced CCL5 production was primarily dependent on Stat1 activation, whereas in CD8+ T cells, Stat1 deficiency does not abrogate CCL5 induction. A chromatin immunoprecipitation assay revealed that in the CCL5 promoter region, both putative Stat3 binding sites exhibit significant binding to Stat3, whereas only one out of four Stat1 binding sites displays moderate binding to Stat1. In tumor-bearing mice, IL-27 induced dramatic production of CCL5 in tumor-infiltrating T cells. IL-27-induced CCL5 appears to contribute to an IL-27-mediated antitumor effect. This is signified by diminished tumor inhibition in anti-CCL5- and IL-27-treated mice. Additionally, intratumor delivery of CCL5 mRNA using lipid nanoparticles significantly inhibited tumor growth. Thus, IL-27 induces robust CCL5 production by T cells, which contributes to antitumor activity.
Collapse
Affiliation(s)
- Aiyan Hu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Jianmin Zhu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Chunxi Zeng
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Cho-Hao Lin
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Jianyu Yu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Jin-Qing Liu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Kimberly Lynch
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Fatemeh Talebian
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Xueliang Pan
- Center for Biostatistics, College of Medicine, The Ohio State University, Columbus, OH
| | - Jingyue Yan
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH; and
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH; and
| | - Zihai Li
- Institute for Immuno-Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Xue-Feng Bai
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH;
- Institute for Immuno-Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| |
Collapse
|
12
|
Monga S, Fares B, Yashaev R, Melamed D, Kahana M, Fares F, Weizman A, Gavish M. The Effect of Natural-Based Formulation (NBF) on the Response of RAW264.7 Macrophages to LPS as an In Vitro Model of Inflammation. J Fungi (Basel) 2022; 8:jof8030321. [PMID: 35330323 PMCID: PMC8955716 DOI: 10.3390/jof8030321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
Macrophages are some of the most important immune cells in the organism and are responsible for creating an inflammatory immune response in order to inhibit the passage of microscopic foreign bodies into the blood stream. Sometimes, their activation can be responsible for chronic inflammatory diseases such as asthma, tuberculosis, hepatitis, sinusitis, inflammatory bowel disease, and viral infections. Prolonged inflammation can damage the organs or may lead to death in serious conditions. In the present study, RAW264.7 macrophages were exposed to lipopolysaccharide (LPS; 20 ng/mL) and simultaneously treated with 20 µg/mL of natural-based formulation (NBF), mushroom–cannabidiol extract). Pro-inflammatory cytokines, chemokines, and other inflammatory markers were analyzed. The elevations in the presence of interleukin-6 (IL-6), cycloxygenase-2 (COX-2), C-C motif ligand-5 (CCL5), and nitrite response, following exposure to LPS, were completely inhibited by NBF administration. IL-1β and tumor necrosis factor alpha (TNF-α) release were inhibited by 3.9-fold and 1.5-fold, respectively. No toxic effect of NBF, as assessed by lactate dehydrogenase (LDH) release, was observed. Treatment of the cells with NBF significantly increased the mRNA levels of TLR2, and TLR4, but not NF-κB. Thus, it appears that the NBF possesses anti-inflammatory and immunomodulatory effects which can attenuate the release of pro-inflammatory markers. NBF may be a candidate for the treatment of acute and chronic inflammatory diseases and deserves further investigation.
Collapse
Affiliation(s)
- Sheelu Monga
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; (S.M.); (F.F.)
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel; (R.Y.); (M.K.)
| | - Basem Fares
- Cannabotech Ltd., 3 Arik Einstein St., Herzliya 4659071, Israel; (B.F.); (D.M.)
| | - Rami Yashaev
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel; (R.Y.); (M.K.)
| | - Dov Melamed
- Cannabotech Ltd., 3 Arik Einstein St., Herzliya 4659071, Israel; (B.F.); (D.M.)
| | - Meygal Kahana
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel; (R.Y.); (M.K.)
| | - Fuad Fares
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; (S.M.); (F.F.)
| | - Abraham Weizman
- Sackler Faculty of Medicine, Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv 6997801, Israel;
- Research Unit, Geha Mental Health Center, Petah Tikva 4910002, Israel
| | - Moshe Gavish
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel; (R.Y.); (M.K.)
- Correspondence:
| |
Collapse
|
13
|
Mehraj U, Mushtaq U, Mir MA, Saleem A, Macha MA, Lone MN, Hamid A, Zargar MA, Ahmad SM, Wani NA. Chemokines in Triple-Negative Breast Cancer Heterogeneity: New Challenges for Clinical Implications. Semin Cancer Biol 2022; 86:769-783. [PMID: 35278636 DOI: 10.1016/j.semcancer.2022.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022]
Abstract
Tumor heterogeneity is a hallmark of cancer and one of the primary causes of resistance to therapies. Triple-negative breast cancer (TNBC), which accounts for 15% to 20% of all breast cancers and is the most aggressive subtype, is very diverse, connected to metastatic potential and response to therapy. It is a very diverse disease at the molecular, pathologic, and clinical levels. TNBC is substantially more likely to recur and has a worse overall survival rate following diagnosis than other breast cancer subtypes. Chemokines, low molecular weight proteins that stimulate chemotaxis, have been shown to control the cues responsible for TNBC heterogeneity. In this review, we have focused on tumor heterogeneity and the role of chemokines in modulating tumor heterogeneity, since this is the most critical issue in treating TNBC. Additionally, we examined numerous cues mediated by chemokine networks that contribute to the heterogeneity of TNBC. Recent developments in our knowledge of the chemokine networks that regulate TNBC heterogeneity may pave the door for developing difficult-to-treat TNBC treatment options.
Collapse
Affiliation(s)
- Umar Mehraj
- Department of Bioresources, School of Life Sciences, University of Kashmir, Srinagar, Jammu & Kashmir India
| | - Umer Mushtaq
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Manzoor A Mir
- Department of Bioresources, School of Life Sciences, University of Kashmir, Srinagar, Jammu & Kashmir India
| | - Afnan Saleem
- Division of Animal Biotechnology Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Sher-e- Kashmir University of Agricultural Sciences and Technology-Kashmir, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science & Technology Awantipora, Jammu & Kashmir, India
| | - Mohammad Nadeem Lone
- Department of Chemistry, School of Physical & Chemical Sciences, Central University of Kashmir, Ganderbal J & K, India
| | - Abid Hamid
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Mohammed A Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Syed Mudasir Ahmad
- Division of Animal Biotechnology Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Sher-e- Kashmir University of Agricultural Sciences and Technology-Kashmir, India
| | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India.
| |
Collapse
|
14
|
Jarboe T, Tuli NY, Chakraborty S, Maniyar RR, DeSouza N, Xiu-Min Li, Moscatello A, Geliebter J, Tiwari RK. Inflammatory Components of the Thyroid Cancer Microenvironment: An Avenue for Identification of Novel Biomarkers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1350:1-31. [PMID: 34888842 DOI: 10.1007/978-3-030-83282-7_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The incidence of thyroid cancer in the United States is on the rise with an appreciably high disease recurrence rate of 20-30%. Anaplastic thyroid cancer (ATC), although rare in occurrence, is an aggressive form of cancer with limited treatment options and bleak cure rates. This chapter uses discussions of in vitro models that are representative of papillary, anaplastic, and follicular thyroid cancer to evaluate the crosstalk between specific cells of the tumor microenvironment (TME), which serves as a highly heterogeneous realm of signaling cascades and metabolism that are associated with tumorigenesis. The cellular constituents of the TME carry out varying characteristic immunomodulatory functions that are discussed throughout this chapter. The aforementioned cell types include cancer-associated fibroblasts (CAFs), endothelial cells (ECs), and cancer stem cells (CSCs), as well as specific immune cells, including natural killer (NK) cells, dendritic cells (DCs), mast cells, T regulatory (Treg) cells, CD8+ T cells, and tumor-associated macrophages (TAMs). TAM-mediated inflammation is associated with a poor prognosis of thyroid cancer, and the molecular basis of the cellular crosstalk between macrophages and thyroid cancer cells with respect to inducing a metastatic phenotype is not yet known. The dynamic nature of the physiological transition to pathological metastatic phenotypes when establishing the TME encompasses a wide range of characteristics that are further explored within this chapter, including the roles of somatic mutations and epigenetic alterations that drive the genetic heterogeneity of cancer cells, allowing for selective advantages that aid in their proliferation. Induction of these proliferating cells is typically accomplished through inflammatory induction, whereby chronic inflammation sets up a constant physiological state of inflammatory cell recruitment. The secretions of these inflammatory cells can alter the genetic makeup of proliferating cells, which can in turn, promote tumor growth.This chapter also presents an in-depth analysis of molecular interactions within the TME, including secretory cytokines and exosomes. Since the exosomal cargo of a cell is a reflection and fingerprint of the originating parental cells, the profiling of exosomal miRNA derived from thyroid cancer cells and macrophages in the TME may serve as an important step in biomarker discovery. Identification of a distinct set of tumor suppressive miRNAs downregulated in ATC-secreted exosomes indicates their role in the regulation of tumor suppressive genes that may increase the metastatic propensity of ATC. Additionally, the high expression of pro-inflammatory cytokines in studies looking at thyroid cancer and activated macrophage conditioned media suggests the existence of an inflammatory TME in thyroid cancer. New findings are suggestive of the presence of a metastatic niche in ATC tissues that is influenced by thyroid tumor microenvironment secretome-induced epithelial to mesenchymal transition (EMT), mediated by a reciprocal interaction between the pro-inflammatory M1 macrophages and the thyroid cancer cells. Thus, targeting the metastatic thyroid carcinoma microenvironment could offer potential therapeutic benefits and should be explored further in preclinical and translational models of human metastatic thyroid cancer.
Collapse
Affiliation(s)
- Tara Jarboe
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Neha Y Tuli
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Sanjukta Chakraborty
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA.,Weill Cornell Medicine, New York, NY, USA
| | - Rachana R Maniyar
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA.,Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicole DeSouza
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Xiu-Min Li
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | | | - Jan Geliebter
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Raj K Tiwari
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
15
|
An Immune-Related Gene Prognostic Index for Triple-Negative Breast Cancer Integrates Multiple Aspects of Tumor-Immune Microenvironment. Cancers (Basel) 2021; 13:cancers13215342. [PMID: 34771505 PMCID: PMC8582543 DOI: 10.3390/cancers13215342] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/17/2021] [Accepted: 10/20/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Triple-negative breast cancer (TNBC) is the most refractory subtype of breast cancer. Immune checkpoint inhibitor (ICI) therapy has made progress in TNBC treatment. PD-L1 expression is a useful biomarker of ICI therapy efficacy. However, tumor-immune microenvironment (TIME) factors, such as immune cell compositions and tumor-infiltrating lymphocyte (TIL) status, also influence tumor immunity. Therefore, it is necessary to seek biomarkers that are associated with multiple aspects of TIME in TNBC. In this study, we developed an immune-related gene prognostic index (IRGPI) with a substantial prognostic value for TNBC. Moreover, the results from multiple cohorts reproducibly demonstrate that IRGPI is significantly associated with immune cell compositions, the exclusion and dysfunction of TILs, as well as PD-1 and PD-L1 expression in TIME. Therefore, IRGPI is a promising biomarker closely related to patient survival and TIME of TNBC and may have a potential effect on the immunotherapy strategy of TNBC. Abstract Tumor-immune cell compositions and immune checkpoints comprehensively affect TNBC outcomes. With the significantly improved survival rate of TNBC patients treated with ICI therapies, a biomarker integrating multiple aspects of TIME may have prognostic value for improving the efficacy of ICI therapy. Immune-related hub genes were identified with weighted gene co-expression network analysis and differential gene expression assay using The Cancer Genome Atlas TNBC data set (n = 115). IRGPI was constructed with Cox regression analysis. Immune cell compositions and TIL status were analyzed with CIBERSORT and TIDE. The discovery was validated with the Molecular Taxonomy of Breast Cancer International Consortium data set (n = 196) and a patient cohort from our hospital. Tumor expression or serum concentrations of CCL5, CCL25, or PD-L1 were determined with immunohistochemistry or ELISA. The constructed IRGPI was composed of CCL5 and CCL25 genes and was negatively associated with the patient’s survival. IRGPI also predicts the compositions of M0 and M2 macrophages, memory B cells, CD8+ T cells, activated memory CD4 T cells, and the exclusion and dysfunction of TILs, as well as PD-1 and PD-L1 expression of TNBC. IRGPI is a promising biomarker for predicting the prognosis and multiple immune characteristics of TNBC.
Collapse
|
16
|
Blockade of Autocrine CCL5 Responses Inhibits Zika Virus Persistence and Spread in Human Brain Microvascular Endothelial Cells. mBio 2021; 12:e0196221. [PMID: 34399621 PMCID: PMC8406327 DOI: 10.1128/mbio.01962-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Zika virus (ZIKV) is a neurovirulent flavivirus that uniquely causes fetal microcephaly, is sexually transmitted, and persists in patients for up to 6 months. ZIKV persistently infects human brain microvascular endothelial cells (hBMECs) that form the blood-brain barrier (BBB) and enables viral spread to neuronal compartments. We found that CCL5, a chemokine with prosurvival effects on immune cells, was highly secreted by ZIKV-infected hBMECs. Although roles for CCL5 in endothelial cell (EC) survival remain unknown, the presence of the CCL5 receptors CCR3 and CCR5 on ECs suggested that CCL5 could promote ZIKV persistence in hBMECs. We found that exogenous CCL5 induced extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation in hBMECs and that ERK1/2 cell survival signaling was similarly activated by ZIKV infection. Neutralizing antibodies to CCL5, CCR3, or CCR5 inhibited persistent ZIKV infection of hBMECs. While knockout (KO) of CCL5 failed to prevent ZIKV infection of hBMECs, at 3 days postinfection (dpi), we observed a >90% reduction in ZIKV-infected CCL5-KO hBMECs and a multilog reduction in ZIKV titers. In contrast, the addition of CCL5 to CCL5-KO hBMECs dose-dependently rescued ZIKV persistence in hBMECs. Inhibiting CCL5 responses using CCR3 (UCB35625) and CCR5 (maraviroc) receptor antagonists reduced the number of ZIKV-infected hBMECs and ZIKV titers (50% inhibitory concentrations [IC50s] of 2.5 to 12 μM), without cytotoxicity (50% cytotoxic concentration [CC50] of >80 μM). These findings demonstrate that ZIKV-induced CCL5 directs autocrine CCR3/CCR5 activation of ERK1/2 survival responses that are required for ZIKV to persistently infect hBMECs. Our results establish roles for CCL5 in ZIKV persistence and suggest the potential for CCL5 receptor antagonists to therapeutically inhibit ZIKV spread and neurovirulence.
Collapse
|
17
|
Chu Y, Nayyar G, Jiang S, Rosenblum JM, Soon-Shiong P, Safrit JT, Lee DA, Cairo MS. Combinatorial immunotherapy of N-803 (IL-15 superagonist) and dinutuximab with ex vivo expanded natural killer cells significantly enhances in vitro cytotoxicity against GD2 + pediatric solid tumors and in vivo survival of xenografted immunodeficient NSG mice. J Immunother Cancer 2021; 9:jitc-2020-002267. [PMID: 34244307 PMCID: PMC8268924 DOI: 10.1136/jitc-2020-002267] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2021] [Indexed: 11/18/2022] Open
Abstract
Background Children with recurrent and/or metastatic osteosarcoma (OS), neuroblastoma (NB) and
glioblastoma multiforme (GBM) have a dismal event-free survival (<25%).
The majority of these solid tumors highly express GD2. Dinutuximab, an anti-GD2
monoclonal antibody, significantly improved event-free survival in children with
GD2+ NB post autologous stem cell transplantation and enhanced natural
killer (NK) cell-mediated antibody-dependent cell cytotoxicity. Thus, approaches to
increase NK cell number and activity, improve persistence and trafficking, and enhance
tumor targeting may further improve the clinical benefit of dinutuximab. N-803 is a
superagonist of an interleukin-15 (IL-15) variant bound to an IL-15 receptor alpha Su-Fc
fusion with enhanced biological activity. Methods The anti-tumor combinatorial effects of N-803, dinutuximab and ex vivo expanded
peripheral blood NK cells (exPBNK) were performed in vitro using cytoxicity assays
against GD2+ OS, NB and GBM cells. Perforin and interferon (IFN)-γ
levels were measured by ELISA assays. Multiple cytokines/chemokines/growth factors
released were measured by multiplex assays. Human OS, GBM or NB xenografted
NOD/SCID/IL2rγnull (NSG) mice were used to investigate the anti-tumor
combinatorial effects in vivo. Results N-803 increased the viability and proliferation of exPBNK. The increased viability and
proliferation are associated with increased phosphorylation of Stat3, Stat5, AKT,
p38MAPK and the expression of NK activating receptors. The combination of dinutuximab
and N-803 significantly enhanced in vitro cytotoxicity of exPBNK with enhanced perforin
and IFN-γ release against OS, GBM and NB. The combination of
exPBNK+N-803+dinutuximab significantly reduced the secretion of tumor necrosis
factor-related apoptosis-inducing ligand (TRAIL), platelet-derived growth factor-BB
(PDGF-BB), and stem cell growth factor beta (SCGF-β) from OS or GBM tumor cells.
Furthermore, OS or GBM significantly inhibited the secretion of regulated on activation,
normal T cell expressed and presumably secreted (RANTES) and stromal cell-derived
factor-1 alpha (SDF-1α) from exPBNK cells (p<0.001) but significantly
enhanced monokine induced by gamma interferon (MIG) secretion from exPBNK cells
(p<0.001). N-803 combined with dinutuximab and exPBNK cells significantly
extended the survival of OS, GBM or NB xenografted NSG mice. Conclusions Our results provide the rationale for the development of a clinical trial of N-803 in
combination with dinutuximab and ex vivo exPBNK cells in patients with recurrent or
metastatic GD2+ solid tumors.
Collapse
Affiliation(s)
- Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Gaurav Nayyar
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Susiyan Jiang
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Jeremy M Rosenblum
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | | | | | - Dean A Lee
- Department of Hem/Onc/BMT, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA .,Department of Medicine, New York Medical College, Valhalla, New York, USA.,Department of Pathology, New York Medical College, Valhalla, New York, USA.,Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, USA.,Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| |
Collapse
|
18
|
Fernández-Nogueira P, Fuster G, Gutierrez-Uzquiza Á, Gascón P, Carbó N, Bragado P. Cancer-Associated Fibroblasts in Breast Cancer Treatment Response and Metastasis. Cancers (Basel) 2021; 13:3146. [PMID: 34201840 PMCID: PMC8268405 DOI: 10.3390/cancers13133146] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BrCa) is the leading cause of death among women worldwide, with about one million new cases diagnosed each year. In spite of the improvements in diagnosis, early detection and treatment, there is still a high incidence of mortality and failure to respond to current therapies. With the use of several well-established biomarkers, such as hormone receptors and human epidermal growth factor receptor-2 (HER2), as well as genetic analysis, BrCa patients can be categorized into multiple subgroups: Luminal A, Luminal B, HER2-enriched, and Basal-like, with specific treatment strategies. Although chemotherapy and targeted therapies have greatly improved the survival of patients with BrCa, there is still a large number of patients who relapse or who fail to respond. The role of the tumor microenvironment in BrCa progression is becoming increasingly understood. Cancer-associated fibroblasts (CAFs) are the principal population of stromal cells in breast tumors. In this review, we discuss the current understanding of CAFs' role in altering the tumor response to therapeutic agents as well as in fostering metastasis in BrCa. In addition, we also review the available CAFs-directed molecular therapies and their potential implications for BrCa management.
Collapse
Affiliation(s)
- Patricia Fernández-Nogueira
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain; (G.F.); (P.G.); (N.C.)
- Department of Biomedicine, School of Medicine, University of Barcelona, 08028 Barcelona, Spain
| | - Gemma Fuster
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain; (G.F.); (P.G.); (N.C.)
- Department of Biochemistry & Physiology, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- Department of Biosciences, Faculty of Sciences and Technology, University of Vic, 08500 Vic, Spain
| | - Álvaro Gutierrez-Uzquiza
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain;
- Health Research Institute of the Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Pere Gascón
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain; (G.F.); (P.G.); (N.C.)
| | - Neus Carbó
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain; (G.F.); (P.G.); (N.C.)
| | - Paloma Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain;
- Health Research Institute of the Hospital Clínico San Carlos, 28040 Madrid, Spain
| |
Collapse
|
19
|
Lu M, Panebra A, Kim WH, Lillehoj HS. Characterization of immunological properties of chicken chemokine CC motif ligand 5 using new monoclonal antibodies. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:104023. [PMID: 33497732 DOI: 10.1016/j.dci.2021.104023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/19/2021] [Accepted: 01/19/2021] [Indexed: 06/12/2023]
Abstract
CCL5 (formerly RANTES) belongs to the CC (or β) chemokine family and is associated with a plethora of inflammatory disorders and pathologic states. CCL5 is mainly produced and secreted by T cells, macrophages, epithelial cells, and fibroblasts and acts as a chemoattractant to recruit effector cells to the inflammation sites. Chicken CCL5 (chCCL5) protein is closely related to avian CCL5 orthologs but distinct from mammalian orthologs, and its modulatory roles in the immune response are largely unknown. The present work was undertaken to characterize the immunological properties of chCCL5 using the new sets of anti-chCCL5 mouse monoclonal antibodies (mAbs). Eight different mAbs (6E11, 6H1, 8H11, 11G1, 11G11, 12H1, 13D1, and 13G3) were characterized for their specificity and binding ability toward chCCL5. Two (13G3 and 6E11) of them were selected to detect native chCCL5 in chCCL5-specific antigen-capture ELISA. Using 13G3 and 6E11 as capture and detection antibodies, respectively, the ELISA system detected serum chCCL5 secretions in Clostridium perfringens- and Eimeria-infected chickens. The intracellular expressions of chCCL5 in primary cells or cell lines derived from chickens were validated in immunocytochemistry and flow cytometry assays using both 13G3 and 6E11 mAbs. Furthermore, 6E11, but not 13G3, neutralized chCCL5-induced chemotaxis in vitro using chicken PBMCs. These molecular characteristics of chCCL5 demonstrate the potential application of anti-chCCL5 mAbs and CCL5-specific antigen-capture detection ELISA for detecting native chCCL5 in biological samples. The availability of these new immunological tools will be valuable for fundamental and applied studies in avian species.
Collapse
Affiliation(s)
- Mingmin Lu
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, 20705, USA
| | - Alfredo Panebra
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, 20705, USA
| | - Woo H Kim
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, 20705, USA
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, 20705, USA.
| |
Collapse
|
20
|
Khan A, Jahejo AR, Qiao ML, Han XY, Cheng QQ, Mangi RA, Qadir MF, Zhang D, Bi YH, Wang Y, Gao GF, Tian WX. NF-кB pathway genes expression in chicken erythrocytes infected with avian influenza virus subtype H9N2. Br Poult Sci 2021; 62:666-671. [PMID: 33843365 DOI: 10.1080/00071668.2021.1902478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
1. Chicken erythrocytes in blood vessels are the most abundant circulating cells, which participate in the host's immune responses. The transcription factor nuclear factor-kappa B (NF-κB) plays a vital role in the inflammatory response following viral infections. However, the expression of the NF-κB pathway, and other immune-related genes in chicken erythrocytes infected with low pathogenic avian influenza virus (LPAIV H9N2), has not been extensively studied.2. The following study determined the interaction of LPAIV H9N2 with chicken erythrocytes using indirect immunofluorescence microscopy. This was followed by investigating myeloid differentiation primary response 88 (MyD88), C-C motif chemokine ligand 5 (CCL5), melanoma differentiation-associated protein 5 (MDA5), the inhibitor of nuclear factor-kappa B kinase subunit epsilon (IKBKE), NF-κB inhibitor alpha (NFKBIA), NF-κB inhibitor epsilon (NFKBIE), interferon-alpha (IFN-α), colony-stimulating factor 3 (CSF3) and tumour necrosis factor receptor-associated factor 6 (TRAF6) by mRNA expression using quantitative real-time PCR (qRT-PCR) at four different time intervals (0, 2, 6 and 10 h).3. There was a significant interaction between erythrocytes and LPAIV H9N2 virus. Furthermore, the mRNA expression of the NF-κB pathway and other immune-related genes were significantly up-regulated at 2 h post-infection in infected chicken erythrocytes, except for TRAF6, which were significantly downregulated. While at 0 h post-infection, IFN-α and CSF3 were significantly upregulated, whereas NFKBIA was significantly downregulated. Further expression of MDA5, CCL5 and NFKBIA was upregulated, while TRAF6 was downregulated at 6 h post-infection. In infected erythrocytes, expression of MyD88, CCL5 and IKBKE was upregulated. However, IFN-α and TRAF6 were downregulated at 10 h post-infection.4. These results give initial evidence that the NF-κB pathway, and other genes related to immunity, in chicken erythrocytes may contribute to LPAIV subtype H9N2 and induce host immune responses.
Collapse
Affiliation(s)
- A Khan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - A R Jahejo
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - M L Qiao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - X Y Han
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Q Q Cheng
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - R A Mangi
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - M F Qadir
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - D Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Y H Bi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Collaborative Innovation Center of Infectious Diseases, Institute of Microbiology, Center for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Science, Beijing, China
| | - Y Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - G F Gao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - W X Tian
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| |
Collapse
|
21
|
Sheng G, Yuan H, Jin L, Ranjit S, Panov J, Lu X, Levi M, Glazer RI. Reduction of fibrosis and immune suppressive cells in ErbB2-dependent tumorigenesis by an LXR agonist. PLoS One 2021; 16:e0248996. [PMID: 33780491 PMCID: PMC8007044 DOI: 10.1371/journal.pone.0248996] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/10/2021] [Indexed: 11/18/2022] Open
Abstract
One of the central challenges for cancer therapy is the identification of factors in the tumor microenvironment that increase tumor progression and prevent immune surveillance. One such element associated with breast cancer is stromal fibrosis, a histopathologic criterion for invasive cancer and poor survival. Fibrosis is caused by inflammatory factors and remodeling of the extracellular matrix that elicit an immune tolerant microenvironment. To address the role of fibrosis in tumorigenesis, we developed NeuT/ATTAC transgenic mice expressing a constitutively active NeuT/erbB2 transgene, and an inducible, fat-directed caspase-8 fusion protein, which upon activation results in selective and partial ablation of mammary fat and its replacement with fibrotic tissue. Induction of fibrosis in NeuT/ATTAC mice led to more rapid tumor development and an inflammatory and fibrotic stromal environment. In an effort to explore therapeutic options that could reduce fibrosis and immune tolerance, mice were treated with the oxysterol liver X receptor (LXR) pan agonist, N,N-dimethyl-3-β-hydroxy-cholenamide (DMHCA), an agent known to reduce fibrosis in non-malignant diseases. DMHCA reduced tumor progression, tumor multiplicity and fibrosis, and improved immune surveillance by reducing infiltrating myeloid-derived suppressor cells and increasing CD4 and CD8 effector T cells. These effects were associated with downregulation of an LXR-dependent gene network related to reduced breast cancer survival that included Spp1, S100a9, Anxa1, Mfge8 and Cd14. These findings suggest that the use of DMHCA may be a potentially effective approach to reduce desmoplasia and immune tolerance and increase the efficacy of cancer therapy.
Collapse
Affiliation(s)
- Gao Sheng
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States of America
- Department of Breast, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Hongyan Yuan
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States of America
| | - Lu Jin
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States of America
| | - Suman Ranjit
- Department of Biochemistry and Molecular Biology, Georgetown University, Washington, DC, United States of America
| | - Julia Panov
- Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Xun Lu
- George Washington University, Washington, DC, United States of America
| | - Moshe Levi
- Department of Biochemistry and Molecular Biology, Georgetown University, Washington, DC, United States of America
| | - Robert I. Glazer
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States of America
- * E-mail:
| |
Collapse
|
22
|
Wang J, Cheng CS, Lu Y, Sun S, Huang S. Volatile Anesthetics Regulate Anti-Cancer Relevant Signaling. Front Oncol 2021; 11:610514. [PMID: 33718164 PMCID: PMC7952859 DOI: 10.3389/fonc.2021.610514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/22/2021] [Indexed: 11/27/2022] Open
Abstract
Volatile anesthetics are widely used inhalation anesthetics in clinical anesthesia. In recent years, the regulation of anti-cancer relevant signaling of volatile anesthetics has drawn the attention of investigators. However, their underlying mechanism remains unclear. This review summarizes the research progress on the regulation of anti-cancer relevant signaling of volatile anesthetics, including sevoflurane, desflurane, xenon, isoflurane, and halothane in vitro, in vivo, and clinical studies. The present review article aims to provide a general overview of regulation of anti-cancer relevant signaling and explore potential underlying molecular mechanisms of volatile anesthetics. It may promote promising insights of guiding clinical anesthesia procedure and instructing enhance recovery after surgery (ERAS) with latent benefits.
Collapse
Affiliation(s)
- Jiaqiang Wang
- Department of Anesthesiology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chien-Shan Cheng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Lu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shen Sun
- Department of Anesthesiology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Shaoqiang Huang
- Department of Anesthesiology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
23
|
Lecchi M, Verderio P, Cappelletti V, De Santis F, Paolini B, Monica M, Sangaletti S, Pupa SM, Iorio MV, Bianchi G, Gennaro M, Fucà G, De Braud F, Tagliabue E, Di Nicola M. A combination of extracellular matrix- and interferon-associated signatures identifies high-grade breast cancers with poor prognosis. Mol Oncol 2021; 15:1345-1357. [PMID: 33523584 PMCID: PMC8096783 DOI: 10.1002/1878-0261.12912] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/14/2020] [Accepted: 01/27/2021] [Indexed: 11/20/2022] Open
Abstract
Breast cancer (BC) is a heterogeneous disease in which the tumor microenvironment (TME) seems to impact the clinical outcome. Here, we investigated whether a combination of gene expression signatures relating to both the structural and immune TME aspects can help predict prognosis in women with high‐grade BC (HGBC). Thus, we focused on a combined molecular biomarker variable that involved extracellular matrix (ECM)‐associated gene expression (ECM3 signature) and interferon (IFN)‐associated metagene (IFN metagene) expression. In 97 chemo‐naive HGBCs from the METABRIC dataset, the dichotomous ECM3/IFN (dECIF) variable identified a group of high‐risk patients (ECM3+/IFN− vs other; hazard ratio = 3.2, 95% confidence interval: 1.5–6.7). Notably, ECM3+/IFN− tumors showed low tumor‐infiltrating lymphocytes, high levels of CD33‐positive cells, absence of PD‐1 expression, or low expression of PD‐L1, as suggested by immune profiles and immune‐histochemical analysis on an independent cohort of 131 HGBCs. To make our results transferable to clinical use, we refined the dECIF biomarker using reduced ECM3 and IFN signatures; notably, the prognostic value of this reduced dECIF was comparable to that of the original dECIF. After validation in a new BC cohort, reduced dECIF was translated into a robust qPCR classifier for real‐world clinical use.
Collapse
Affiliation(s)
- Mara Lecchi
- Bioinformatics and Biostatistics Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Verderio
- Bioinformatics and Biostatistics Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Vera Cappelletti
- Biomarker Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca De Santis
- Unit of Immunotherapy and Anticancer Innovative Therapeutics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Biagio Paolini
- Pathology A Unit, Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Melissa Monica
- Pathology A Unit, Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sabina Sangaletti
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Serenella Maria Pupa
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marilena Valeria Iorio
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Bianchi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Massimiliano Gennaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanni Fucà
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo De Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Massimo Di Nicola
- Unit of Immunotherapy and Anticancer Innovative Therapeutics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
24
|
Anti-diabetic drug metformin inhibits cell proliferation and tumor growth in gallbladder cancer via G0/G1 cell cycle arrest. Anticancer Drugs 2021; 31:231-240. [PMID: 31815765 DOI: 10.1097/cad.0000000000000870] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gallbladder cancer is the most common biliary tract cancer with poor prognosis and wide variation in incidence rates worldwide, being very high in some countries in Latin America and Asia. Treatment of type 2 diabetes with metformin causes a reduction in the incidence of cancer. Till date, there are no reports on the anti-tumor effects of metformin in gall bladder cancer. Therefore, this study evaluated the effects of metformin on the proliferation of human gallbladder adenocarcinoma cells in vitro and in vivo, as well as explored the microRNAs associated with the anti-tumor effects of metformin. Metformin inhibited the proliferation in gallbladder adenocarcinoma cell lines NOZ, TGBC14TKB, and TGBC24TKB, and blocked the G0 to G1 transition in the cell cycle. This was accompanied by strong reduction in the expression of G1 cyclins, especially cyclin D1 and its catalytic subunits including cyclin-dependent kinase 4, and in retinoblastoma protein phosphorylation. In addition, metformin reduced the phosphorylation of receptor tyrosine kinases, especially Tie-2, ALK, PYK, EphA4, and EphA10, as well as angiogenesis-related proteins, including RANTES, TGF-β, and TIMP-1. Moreover, metformin also markedly altered microRNA expression profile leading to an anti-tumor effect. Treatment of athymic nude mice bearing xenograft tumors with metformin inhibited tumor growth. These results suggest that metformin may be used clinically for the treatment of gallbladder adenocarcinoma.
Collapse
|
25
|
Ghandhariyoun N, Jaafari MR, Nikoofal-Sahlabadi S, Taghdisi SM, Moosavian SA. Reducing Doxorubicin resistance in breast cancer by liposomal FOXM1 aptamer: In vitro and in vivo. Life Sci 2020; 262:118520. [PMID: 33010284 DOI: 10.1016/j.lfs.2020.118520] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 12/14/2022]
Abstract
AIMS Drug resistance is one of the main obstacles in cancer chemotherapy. The forkhead box M1 (FOXM1) is a transcription factor and its overexpression in breast cancer is related to resistance to chemotherapy. In this study, we prepare liposomal FOXM1 aptamer (Lip-FOXM1apt) and evaluate its effects on Doxorubicin (Dox) resistance in vitro and in vivo. MAIN METHODS MTT assay, cell association, cellular uptake, Annexin V-FITC/PI dual staining assay were investigated in MDA-MB-231, MCF-7, 4T1. In vivo studies were performed in 4T1 tumor-bearing BALB/c mice. KEY FINDINGS We found that the combination therapy of Dox and Lip-FOXM1apt significantly increases both Dox cytotoxicity on cancer cells as well as Dox-induced apoptosis. Administering Lip-FOXM1apt remarkably improved the anti-tumor efficacy of Dox in mice model that was strikingly more effective than Dox monotherapy. SIGNIFICANCE Taken together, this study provides a new strategy to overcome Dox resistance and merits further investigation.
Collapse
Affiliation(s)
- Negin Ghandhariyoun
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, P.O. Box 91775-1365, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, P.O. Box 91775-1365, Mashhad, Iran
| | - Sara Nikoofal-Sahlabadi
- Department of Pharmaceutics, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Alia Moosavian
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, P.O. Box 91775-1365, Mashhad, Iran.
| |
Collapse
|
26
|
Water-Pipe Smoking Exposure Deregulates a Set of Genes Associated with Human Head and Neck Cancer Development and Prognosis. TOXICS 2020; 8:toxics8030073. [PMID: 32961854 PMCID: PMC7560251 DOI: 10.3390/toxics8030073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/31/2022]
Abstract
Water-pipe smoking (WPS) is becoming the most popular form of tobacco use among the youth, especially in the Middle East, replacing cigarettes rapidly and becoming a major risk of tobacco addiction worldwide. Smoke from WPS contains similar toxins as those present in cigarette smoke and is linked directly with different types of cancers including lung and head and neck (HN) carcinomas. However, the underlying molecular pathways and/or target genes responsible for the carcinogenic process are still unknown. In this study, human normal oral epithelial (HNOE) cells, NanoString PanCancer Pathways panel of 770 gene transcripts and quantitative real-time polymerase chain reaction (qRT-PCR) analysis were applied to discover differentially expressed genes (DEG) modulated by WPS. In silico analysis was performed to analyze the impact of these genes in HN cancer patient’s biology and outcome. We found that WPS can induce the epithelial–mesenchymal transition (EMT: hallmark of cancer progression) of HNOE cells. More significantly, our analysis of NanoString revealed 23 genes deregulated under the effect of WPS, responsible for the modulation of cell cycle, proliferation, migration/invasion, apoptosis, signal transduction, and inflammatory response. Further analysis was performed using qRT-PCR of HNOE WPS-exposed and unexposed cells supported the reliability of our NanoString data. Moreover, we demonstrate those DEG to be upregulated in cancer compared with normal tissue. Using the Kaplan–Meier analysis, we observed a significant association between WPS-deregulated genes and relapse-free survival/overall survival in HN cancer patients. Our findings imply that WPS can modulate EMT as well as a set of genes that are directly involved in human HN carcinogenesis, thereby affecting HN cancer patients’ survival.
Collapse
|
27
|
The CCL5/CCR5 Axis in Cancer Progression. Cancers (Basel) 2020; 12:cancers12071765. [PMID: 32630699 PMCID: PMC7407580 DOI: 10.3390/cancers12071765] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor cells can “hijack” chemokine networks to support tumor progression. In this context, the C-C chemokine ligand 5/C-C chemokine receptor type 5 (CCL5/CCR5) axis is gaining increasing attention, since abnormal expression and activity of CCL5 and its receptor CCR5 have been found in hematological malignancies and solid tumors. Numerous preclinical in vitro and in vivo studies have shown a key role of the CCL5/CCR5 axis in cancer, and thus provided the rationale for clinical trials using the repurposed drug maraviroc, a CCR5 antagonist used to treat HIV/AIDS. This review summarizes current knowledge on the role of the CCL5/CCR5 axis in cancer. First, it describes the involvement of the CCL5/CCR5 axis in cancer progression, including autocrine and paracrine tumor growth, ECM (extracellular matrix) remodeling and migration, cancer stem cell expansion, DNA damage repair, metabolic reprogramming, and angiogenesis. Then, it focuses on individual hematological and solid tumors in which CCL5 and CCR5 have been studied preclinically. Finally, it discusses clinical trials of strategies to counteract the CCL5/CCR5 axis in different cancers using maraviroc or therapeutic monoclonal antibodies.
Collapse
|
28
|
Liu S, Zhou X, Peng X, Li M, Ren B, Cheng G, Cheng L. Porphyromonas gingivalis Promotes Immunoevasion of Oral Cancer by Protecting Cancer from Macrophage Attack. THE JOURNAL OF IMMUNOLOGY 2020; 205:282-289. [PMID: 32471882 DOI: 10.4049/jimmunol.1901138] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 04/21/2020] [Indexed: 02/05/2023]
Abstract
The relationship of Porphyromonas gingivalis and oral squamous cell carcinoma (OSCC) has been studied for several years. Previous studies have focused on the direct effect of P. gingivalis on the activities of primary epithelial cells and OSCC cells. However, the immune system is responsible for mediating cancer development, whether P. gingivalis can affect oral cancer immunity has seldom been explored to date. In this study, we investigated the role of P. gingivalis in the immunoevasion of OSCC. We evaluated the effect of P. gingivalis on the phagocytosis of Cal-27 cells (OSCC cell line) by bone marrow-derived macrophages in vitro and studied the effect of P. gingivalis on the growth of OSCC and the polarization of tumor-associated macrophages in vivo. We found that P. gingivalis was able to inhibit the phagocytosis of Cal-27 cells by macrophages, and membrane-component molecules of P. gingivalis, such as proteins, were speculated to be the effector components. In addition, sustained infection with antibiotics-inactivated P. gingivalis promoted OSCC growth in mice and induced the polarization of macrophages into M2 tumor-associated macrophages, which mainly display protumor properties. Transcriptome analysis and quantitative RT-PCR revealed that P. gingivalis infection upregulated the expression of genes encoding protumor molecules in Cal-27 cells (suprabasin, IL-1R2, and CD47) and in macrophages (IL-1α, CCL-3, and CCL-5). Our in vitro and in vivo data suggest that P. gingivalis can promote immunoevasion of oral cancer by protecting cancer from macrophage attack. To our knowledge, the present study reveals a novel mechanism by which P. gingivalis promotes OSCC development.
Collapse
Affiliation(s)
- Shiyu Liu
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.,National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China
| | - Xian Peng
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China.,National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China
| | - Mingyun Li
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China.,National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China.,National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China
| | - Guo Cheng
- Laboratory of Molecular Translational Medicine, Centre for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610017, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China; .,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.,National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China
| |
Collapse
|
29
|
Phutim-Mangkhalthon A, Teerakapong A, Tippayawat P, Morales NP, Morkmued S, Puasiri S, Priprem A, Damrongrungruang T. Anti-inflammatory effect of photodynamic therapy using guaiazulene and red lasers on peripheral blood mononuclear cells. Photodiagnosis Photodyn Ther 2020; 31:101747. [PMID: 32200021 DOI: 10.1016/j.pdpdt.2020.101747] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/18/2020] [Accepted: 03/16/2020] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Photodynamic therapy improves oral mucositis treatment. The reactive oxygen species (ROS) generated from this reaction could contribute to an anti-inflammatory effect by suppressing inflammatory cells. OBJECTIVE To evaluate the anti-inflammatory effect of photodynamic therapy using guaiazulene and a red laser in peripheral blood mononuclear cells (PBMCs). METHODS Guaiazulene solutions (1, 2, 5, 25, 35, and 100 μM in 99.8 % methanol) were irradiated with red laser light (625 nm, 146.2 mW/cm2) in continuous mode at 0, 4, and 8 J/cm2 in black 96-well plates. ROS were measured using spin trapping technique with electron spin resonance (ESR) spectroscopy and fluorescence. The two highest concentrations were tested using cell viability (PrestoBlue®) and anti-inflammation (RANTES and PGE2 ELISA) assay kits. Kruskal-Wallis and Dunn Bonferroni tests were used for statistical analyses with significant differences at p-value < 0.05. RESULTS Guaiazulene solutions between 2 and 5 μM exposed to red laser light at 4-8 J/cm2 generated significantly more singlet oxygen compared to the no guaiazulene group (p < 0.01) and reduced RANTES and PGE2 levels in TNF-α-inflamed peripheral blood mononuclear cells without affecting cell viability. CONCLUSION Photodynamic activation of guaiazulene generated singlet oxygen and suppressed inflammatory markers in PBMCs.
Collapse
Affiliation(s)
- Ampika Phutim-Mangkhalthon
- Division of Paediatric Dentistry, Department of Preventive Dentistry, Faculty of Dentistry, Khon Kaen University, 40002, Thailand.
| | - Aroon Teerakapong
- Division of Periodontology, Department of Oral Biomedical Sciences, Faculty of Dentistry, Khon Kaen University, 40002, Thailand; Lasers in Dentistry Research Group, Khon Kaen University, Thailand
| | - Patcharaporn Tippayawat
- Department of Clinical Chemistry, Faculty of Associated Medical Sciences, Khon Kaen University, 40002, Thailand.
| | | | - Supawich Morkmued
- Division of Paediatric Dentistry, Department of Preventive Dentistry, Faculty of Dentistry, Khon Kaen University, 40002, Thailand.
| | - Subin Puasiri
- Department of Dental Public Health, Faculty of Dentistry, Khon Kaen University, 40002, Thailand.
| | - Aroonsri Priprem
- Melatonin Research Group and Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand.
| | - Teerasak Damrongrungruang
- Division of Oral Diagnosis, Department of Oral Biomedical Science, Faculty of Dentistry, Khon Kaen University, 40002, Thailand; Lasers in Dentistry Research Group, Khon Kaen University, Thailand.
| |
Collapse
|
30
|
Shan J, Chouchane A, Mokrab Y, Saad M, Boujassoum S, Sayaman RW, Ziv E, Bouaouina N, Remadi Y, Gabbouj S, Roelands J, Ma X, Bedognetti D, Chouchane L. Genetic Variation in CCL5 Signaling Genes and Triple Negative Breast Cancer: Susceptibility and Prognosis Implications. Front Oncol 2019; 9:1328. [PMID: 31921621 PMCID: PMC6915105 DOI: 10.3389/fonc.2019.01328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/13/2019] [Indexed: 12/17/2022] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for ~15–20% of breast cancer (BC) and has a higher rate of early relapse and mortality compared to other subtypes. The Chemokine (C-C motif) ligand 5 (CCL5) and its signaling pathway have been linked to TNBC. We aimed to investigate the susceptibility and prognostic implications of genetic variation in CCL5 signaling genes in TNBC in the present study. We characterized variants in CCL5 and that of six other CCL5 signaling genes (CCND1, ZMIZ1, CASP8, NOTCH2, MAP3K21, and HS6ST3) among 1,082 unrelated Tunisian subjects (544 BC patients, including 196 TNBC, and 538 healthy controls), assessed the association of the variants with BC-specific overall survival (OVS) and progression-free survival (PFS), and correlated CCL5 mRNA and serum levels with CCL5 genotypes. We found a highly significant association between the CCND1 rs614367-TT genotype (OR = 5.14; P = 0.004) and TNBC risk, and identified a significant association between the rs614367-T allele and decreased PFS in TNBC. A decreased risk of lymph node metastasis was associated with the MAP3K21 rs1294255-C allele, particularly in rs1294255-GC (OR = 0.47; P = 0.001). CCL5 variants (rs2107538 and rs2280789) were linked to CCL5 serum and mRNA levels. In the TCGA TNBC/Basal-like cohort the MAP3K21 rs1294255-G allele was associated with a decreased OVS. High expression of CCL5 in breast tumors was significantly associated with an increased OVS in all BC patients, but particularly in TNBC/Basal-like patients. In conclusion, genetic variation in CCL5 signaling genes may predict not only TNBC risk but also disease aggressiveness.
Collapse
Affiliation(s)
- Jingxuan Shan
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, United States.,Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, United States.,Laboratory of Genetic Medicine and Immunology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Aziz Chouchane
- Faculta di Medicina e Chirurgia, Universita Cattolica del Sacro Cuero, Rome, Italy
| | - Younes Mokrab
- Translational Genetics and Bioinformatics Section, Research Division, Sidra Medicine, Doha, Qatar
| | - Mohamad Saad
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Salha Boujassoum
- Department of Medical Oncology, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Rosalyn W Sayaman
- Department of Population Sciences, City of Hope, Duarte, CA, United States.,Department of Laboratory Medicine at UCSF, San Francisco, CA, United States.,Helen Diller Family Comprehensive Cancer Center at UCSF, San Francisco, CA, United States
| | - Elad Ziv
- Helen Diller Family Comprehensive Cancer Center at UCSF, San Francisco, CA, United States.,Division of General Internal Medicine, Department of Medicine, Institute for Human Genetics at UCSF, San Francisco, CA, United States
| | - Noureddine Bouaouina
- Service de Cancérologie Radiothérapie, CHU Farhat Hached, Sousse, Tunisia.,Laboratoire d'Immuno-Oncologie Moléculaire, Faculté de Médecine de Monastir, Université de Monastir, Monastir, Tunisia
| | - Yasmine Remadi
- Laboratoire d'Immuno-Oncologie Moléculaire, Faculté de Médecine de Monastir, Université de Monastir, Monastir, Tunisia
| | - Sallouha Gabbouj
- Laboratoire d'Immuno-Oncologie Moléculaire, Faculté de Médecine de Monastir, Université de Monastir, Monastir, Tunisia
| | - Jessica Roelands
- Tumor Biology Section, Research Division, Sidra Medicine, Doha, Qatar
| | - Xiaojing Ma
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, United States
| | - Davide Bedognetti
- Tumor Biology Section, Research Division, Sidra Medicine, Doha, Qatar
| | - Lotfi Chouchane
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, United States.,Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, United States.,Laboratory of Genetic Medicine and Immunology, Weill Cornell Medicine-Qatar, Doha, Qatar
| |
Collapse
|
31
|
Voshtani R, Song M, Wang H, Li X, Zhang W, Tavallaie MS, Yan W, Sun J, Wei F, Ma X. Progranulin promotes melanoma progression by inhibiting natural killer cell recruitment to the tumor microenvironment. Cancer Lett 2019; 465:24-35. [PMID: 31491449 DOI: 10.1016/j.canlet.2019.08.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/23/2019] [Accepted: 08/30/2019] [Indexed: 12/20/2022]
Abstract
Progranulin (PGRN) is a growth factor with significant biological effects in different types of cancer. However, its role in melanoma progression has not been explored. In this study, we first analyze clinical datasets and show that high PGRN expression levels are correlated with poor prognosis of melanoma patients. Further, we demonstrate in a transplanted murine melanoma model in which the endogenous Grn gene encoding PGRN has been deleted that tumor-derived, not host-derived PGRN, promotes melanoma growth and metastasis. Immunological analyses reveal an enhanced infiltration of natural killer cells, but not T lymphocytes, into PGRN-deficient tumors compared to the wild type control. Antibody-mediated depletion confirms the critical role of NK cells in controlling B16 tumor growth. RNA-seq analysis reveals that several chemokines including CCL5 are strongly upregulated in PGRN-deficient tumor. Silencing CCL5 expression in PGRN-deficient tumor reduces NK cell recruitment and restores tumor growth to the control level. Lastly, we show that PGRN inhibits Ccl5 gene expression at the transcriptional level. This study highlights a novel and critical role of PGRN in melanoma growth and metastasis and suggests that it may represent a potential therapeutic target.
Collapse
Affiliation(s)
- Ramouna Voshtani
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, China
| | - Mei Song
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, USA
| | - Huan Wang
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, China
| | - Xiaoqi Li
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, China
| | - Wei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mojdeh S Tavallaie
- Department of Pharmaceutical Sciences, Shanghai Jiaotong University, Shanghai, 200240, China
| | - Wenjun Yan
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, China
| | - Joseph Sun
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Fang Wei
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, China.
| | - Xiaojing Ma
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, China; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, USA.
| |
Collapse
|
32
|
Nabizadeh JA, Manthey HD, Panagides N, Steyn FJ, Lee JD, Li XX, Akhir FNM, Chen W, Boyle GM, Taylor SM, Woodruff TM, Rolfe BE. C5a receptors C5aR1 and C5aR2 mediate opposing pathologies in a mouse model of melanoma. FASEB J 2019; 33:11060-11071. [PMID: 31298935 DOI: 10.1096/fj.201800980rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The canonical complement component 5a (C5a) receptor (C5aR) 1 has well-described roles in tumorigenesis but the contribution of the second receptor, C5aR2, is unclear. The present study demonstrates that B16.F0 melanoma cells express mRNA for both C5aR1 and C5aR2 and signal through ERK and p38 MAPKs in response to C5a. Despite this, C5a had no impact on melanoma cell proliferation or migration in vitro. In vivo studies demonstrated that the growth of B16.F0 melanoma tumors was increased in C5aR2-/- mice but reduced in C5aR1-/- mice and wild-type mice treated with a C5aR1 antagonist. Analysis of tumor-infiltrating leukocyte populations showed no significant differences between wild-type and C5aR2-/- mice. Conversely, percentages of myeloid-derived suppressor cells, macrophages, and regulatory T lymphocytes were lower in tumors from C5aR1-/- mice, whereas total (CD3+) T lymphocytes and CD4+ subsets were higher. Analysis of cytokine and chemokine levels also showed plasma IFN-γ was higher and tumor C-C motif chemokine ligand 2 was lower in the absence of C5aR1. The results suggest that C5aR1 signaling supports melanoma growth by promoting infiltration of immunosuppressive leukocyte populations into the tumor microenvironment, whereas C5aR2 has a more restricted but beneficial role in limiting tumor growth. Overall, these data support the potential of C5aR1-inhibitory therapies for melanoma.-Nabizadeh, J. A., Manthey, H. D., Panagides, N., Steyn, F. J., Lee, J. D., Li, X. X., Akhir, F. N. M., Chen, W., Boyle, G. M., Taylor, S. M., Woodruff, T. M., Rolfe, B. E. C5a receptors C5aR1 and C5aR2 mediate opposing pathologies in a mouse model of melanoma.
Collapse
Affiliation(s)
- Jamileh A Nabizadeh
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Helga D Manthey
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Nadya Panagides
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Frederik J Steyn
- Centre for Clinical Research, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - John D Lee
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Xaria X Li
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Fazrena N M Akhir
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Weiyu Chen
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Glen M Boyle
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Stephen M Taylor
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Trent M Woodruff
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Barbara E Rolfe
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
33
|
Turbitt WJ, Xu Y, Sosnoski DM, Collins SD, Meng H, Mastro AM, Rogers CJ. Physical Activity Plus Energy Restriction Prevents 4T1.2 Mammary Tumor Progression, MDSC Accumulation, and an Immunosuppressive Tumor Microenvironment. Cancer Prev Res (Phila) 2019; 12:493-506. [PMID: 31266827 DOI: 10.1158/1940-6207.capr-17-0233] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/09/2017] [Accepted: 06/26/2019] [Indexed: 11/16/2022]
Abstract
Physical activity and the prevention of weight gain decrease breast cancer incidence and improve survival. Unraveling the biological mechanisms underlying these cancer prevention effects is difficult because activity and dietary restriction are often linked. The goal of this study was to determine whether physical activity (PA), preventing weight gain via energy restriction (ER), or the combination was most effective in delaying tumor growth, reducing metastatic progression, and improving survival in the 4T1.2 mammary tumor model. Furthermore, we determined whether any of these interventions prevented the expansion of protumor immunosuppressive cells and altered the tumor microenvironment (TME). Female BALB/c mice (n = 7-20/group) were randomized to sedentary (SED) or PA wheel cages and fed ad libitum (AL) or 90% of control food intake (ER). After 8 weeks on the interventions, mice were inoculated with 5 × 104 4T1.2luc cells into the 4th mammary fat pad and continued on their respective intervention. PA+ER significantly delayed primary tumor growth (final tumor volume, 0.193 ± 0.042 vs. 0.369 ± 0.049 cm3, P < 0.001), reduced metastatic burden in the lungs (0.72 ± 0.36 vs. 16.27 ± 6.98, P = 0.054) and increased survival (median survival, 68 vs 40 days, P = 0.043) compared with SED+AL mice. PA+ER also reduced the expression level of metastatic and immunosuppressive genes and resulted in favorable changes in immune cell infiltrates in the tumor. These data suggest that both PA and ER are needed to reduce tumor growth, delay metastatic progression, and improve survival, and that this protection is associated with changes in immune-mediated mechanisms.
Collapse
Affiliation(s)
- William J Turbitt
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania
| | - Yitong Xu
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania
| | - Donna M Sosnoski
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania
| | - Shawntawnee D Collins
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania
| | - Huicui Meng
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania
| | - Andrea M Mastro
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania.,Penn State Hershey Cancer Institute, Hershey, Pennsylvania
| | - Connie J Rogers
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania. .,Penn State Hershey Cancer Institute, Hershey, Pennsylvania.,Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
34
|
Ullah M, Akbar A, Ng NN, Concepcion W, Thakor AS. Mesenchymal stem cells confer chemoresistance in breast cancer via a CD9 dependent mechanism. Oncotarget 2019; 10:3435-3450. [PMID: 31191817 PMCID: PMC6544397 DOI: 10.18632/oncotarget.26952] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/05/2019] [Indexed: 12/11/2022] Open
Abstract
The development of chemotherapy drug resistance remains a significant barrier for effective therapy in several cancers including breast cancer. Bone marrow-derived mesenchymal stem cells (BMMSCs) have previously been shown to influence tumor progression and the development of chemoresistance. In the present study, we showed that when GFP labelled BMMSCs and RFP labelled HCC1806 cells are injected together in vivo, they create tumors which contain a new hybrid cell that has characteristics of both BMMSCs and HCC1806 cells. By labelling these cells prior to their injection, we were then able to isolate new hybrid cell from harvested tumors using FACS (DP-HCC1806:BMMSCs). Interestingly, when DP-HCC1806:BMMSCs were then injected into the mammary fat pad of NOD/SCID mice, they produced xenograft tumors which were smaller in size, and exhibited resistance to chemotherapy drugs (i.e. doxorubicin and 5-fluorouracil), when compared tumors from HCC1806 cells alone. This chemoresistance was shown to associated with an increased expression of tetraspanins (CD9, CD81) and drug resistance proteins (BCRP, MDR1). Subsequent siRNA-mediated knockdown of BMMSC-CD9 in DP-HCC1806:BMMSCs resulted in an attenuation of doxorubicin and 5-fluorouracil chemoresistance associated with decreased BCRP and serum cytokine expression (CCL5, CCR5, CXCR12). Our findings suggest that within the tumor microenvironment, CD9 is responsible for the crosstalk between BMMSCs and HCC1806 breast cancer cells (via CCL5, CCR5, and CXCR12) which contributes to chemoresistance. Hence, BMMSC-CD9 may serve as an important therapeutic target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, CA 94304, USA
| | - Asma Akbar
- Mid-Florida Research and Education Center, Department of Pathology, University of Florida, Apopka, FL 32703, USA
| | - Nathan Norton Ng
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, CA 94304, USA
| | - Waldo Concepcion
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, CA 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, CA 94304, USA
| |
Collapse
|
35
|
Shan J, Li S, Wang C, Liu L, Wang X, Zhu D, Fan Y, Xu J. Expression and biological functions of the CCL5‐CCR5 axis in oral lichen planus. Exp Dermatol 2019; 28:816-821. [PMID: 31006151 DOI: 10.1111/exd.13946] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 04/02/2019] [Accepted: 04/14/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Jing Shan
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Shan Li
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
- Department of Stomatology Yancheng City No. 1 People's Hospital Yancheng China
| | - Chen Wang
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Lin Liu
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Xuewei Wang
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Dandan Zhu
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Yuan Fan
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Juanyong Xu
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| |
Collapse
|
36
|
Identification of a novel biomarker-CCL5 using antibody microarray for colorectal cancer. Pathol Res Pract 2019; 215:1033-1037. [DOI: 10.1016/j.prp.2019.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 01/25/2019] [Accepted: 02/26/2019] [Indexed: 01/04/2023]
|
37
|
Pydi SS, Ghousunnissa S, Devalraju KP, Ramaseri SS, Gaddam R, Auzumeedi SK, Vankayalapati R, Valluri VL. Down regulation of RANTES in pleural site is associated with inhibition of antigen specific response in tuberculosis. Tuberculosis (Edinb) 2019; 116S:S123-S130. [PMID: 31103419 DOI: 10.1016/j.tube.2019.04.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 11/21/2018] [Indexed: 12/17/2022]
Abstract
Tuberculosis is the most common infectious reason for death and a major cause of pleural effusion globally. To understand the role of chemokines in trafficking of cells during TB pleurisy, we studied the responses to MTB, Ag85A in cells from pleural fluids and peripheral blood. Patients with TB pleural effusions, malignant effusions and asymptomatic healthy controls were enrolled. High expression (p < 0.05) of IP-10, MCP-1, MIG, IL-8, IFN-γ and IL-23 were observed in pleural fluids of TB patients compared to their plasma where expression of RANTES was significantly higher (p < 0.05). On specific stimulation of PFMCs with Ag85A, expression of RANTES was significantly lower in TB compared to NTB patients. We also observed increased expression of T regs and PD1 on CD8+T cells in PFMC of TB patients. Though some of the inflammatory chemokine/cytokines were up-regulated in pleura of TB patients, antigenic stimulation failed to induce them indicating poor antigenic responses at the site. Low expression of RANTES might be a reason for decreased trafficking of cells to the site and dissemination of infection into pleural site. The pattern of RANTES expression in pleural fluid vs serum is interesting. The observations necessitate further studies to investigate the levels of RANTES for its potential biological relevance in TB immunity and its use as a biomarker for diagnosis of pleural TB.
Collapse
Affiliation(s)
- Satya Sudheer Pydi
- Immunology and Molecular Biology Division, Blue Peter Public Health and Research Centre, LEPRA Society, Cherlapally, Hyderabad, 501301, India
| | - Sheikh Ghousunnissa
- Immunology and Molecular Biology Division, Blue Peter Public Health and Research Centre, LEPRA Society, Cherlapally, Hyderabad, 501301, India
| | - Kamakshi Prudhula Devalraju
- Immunology and Molecular Biology Division, Blue Peter Public Health and Research Centre, LEPRA Society, Cherlapally, Hyderabad, 501301, India
| | - Sharadambal Sunder Ramaseri
- Immunology and Molecular Biology Division, Blue Peter Public Health and Research Centre, LEPRA Society, Cherlapally, Hyderabad, 501301, India
| | - Ramulu Gaddam
- Department of Respiratory Medicine, AP Chest and General Hospital, Osmania Medical College, Erragadda, Hyderabad, 500038, India
| | - Sai Kumar Auzumeedi
- Department of Respiratory Medicine, AP Chest and General Hospital, Osmania Medical College, Erragadda, Hyderabad, 500038, India
| | - Ramakrishna Vankayalapati
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Center, Tyler, TX, 75708, USA.
| | - Vijaya Lakshmi Valluri
- Immunology and Molecular Biology Division, Blue Peter Public Health and Research Centre, LEPRA Society, Cherlapally, Hyderabad, 501301, India.
| |
Collapse
|
38
|
Liubomirski Y, Lerrer S, Meshel T, Morein D, Rubinstein-Achiasaf L, Sprinzak D, Wiemann S, Körner C, Ehrlich M, Ben-Baruch A. Notch-Mediated Tumor-Stroma-Inflammation Networks Promote Invasive Properties and CXCL8 Expression in Triple-Negative Breast Cancer. Front Immunol 2019; 10:804. [PMID: 31105691 PMCID: PMC6492532 DOI: 10.3389/fimmu.2019.00804] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 03/26/2019] [Indexed: 01/12/2023] Open
Abstract
Stromal cells and pro-inflammatory cytokines play key roles in promoting the aggressiveness of triple-negative breast cancers (TNBC; Basal/Basal-like). In our previous study we demonstrated that stimulation of TNBC and mesenchymal stem cells (MSCs) co-cultures by the pro-inflammatory cytokine tumor necrosis factor α (TNFα) has led to increased metastasis-related properties in vitro and in vivo. In this context, elevated release of the pro-metastatic chemokines CXCL8 (IL-8) and CCL5 (RANTES) was noted in TNFα- and interleukin-1β (IL-1β)-stimulated TNBC:MSC co-cultures; the process was partly (CXCL8) and entirely (CCL5) dependent on physical contacts between the two cell types. Here, we demonstrate that DAPT, inhibitor of γ-secretase that participates in activation of Notch receptors, inhibited the migration and invasion of TNBC cells that were grown in “Contact” co-cultures with MSCs or with patient-derived cancer-associated fibroblasts (CAFs), in the presence of TNFα. DAPT also inhibited the contact-dependent induction of CXCL8, but not of CCL5, in TNFα- and IL-1β-stimulated TNBC:MSC/CAF co-cultures; some level of heterogeneity between the responses of different TNBC cell lines was noted, with MDA-MB-231:MSC/CAF co-cultures being the most sensitive to DAPT. Patient dataset studies comparing basal tumors to luminal-A tumors, and mRNA analyses of Notch receptors in TNBC and luminal-A cells pointed at Notch1 as possible mediator of CXCL8 increase in TNFα-stimulated TNBC:stroma “Contact” co-cultures. Accordingly, down-regulation of Notch1 in TNBC cells by siRNA has substantially reduced the contact-dependent elevation in CXCL8 in TNFα- and also in IL-1β-stimulated TNBC:MSC “Contact” co-cultures. Then, studies in which CXCL8 or p65 (NF-κB pathway) were down-regulated (siRNAs; CRISPR/Cas9) in TNBC cells and/or MSCs, indicated that upon TNFα stimulation of “Contact” co-cultures, p65 was activated and led to CXCL8 production mainly in TNBC cells. Moreover, our findings indicated that when tumor cells interacted with stromal cells in the presence of pro-inflammatory stimuli, TNFα-induced p65 activation has led to elevated Notch1 expression and activation, which then gave rise to elevated production of CXCL8. Overall, tumor:stroma interactions set the stage for Notch1 activation by pro-inflammatory signals, leading to CXCL8 induction and consequently to pro-metastatic activities. These observations may have important clinical implications in designing novel therapy combinations in TNBC.
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
| | - Dina Morein
- 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
| | - David Sprinzak
- School of Neurobiology, Biochemistry & Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cindy Körner
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcelo Ehrlich
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Adit Ben-Baruch
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
39
|
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
|
40
|
CC chemokines are differentially expressed in Breast Cancer and are associated with disparity in overall survival. Sci Rep 2019; 9:4014. [PMID: 30850664 PMCID: PMC6408438 DOI: 10.1038/s41598-019-40514-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/18/2019] [Indexed: 12/21/2022] Open
Abstract
Despite recent advances, breast cancer (BrCa) still affects many women and the impact is disproportional in African Americans (AA) compared to European Americans (EA). Addressing socioeconomic and behavioral status has not been enough to reduce disparity, suggesting contribution of biological differences in BrCa disparity. Our laboratory was first to show involvement of CC chemokines in BrCa. In this study, using ONCOMINE, TCGA, bc-GenExMiner and KMplotter, we examined the association of CC chemokines in BrCa outcomes and disparity. We show over-expression of CCL5, -7, -11, -17, -20, -22 and -25 in BrCa tissues. High mRNA levels of CCL7, -8, -17, -20 and -25 predicted a decrease in overall survival (OS). CCL7 and CCL8 were associated with decreased relapse-free survival. Expression of CCL17 and CCL25 was associated with decreased OS in AA. In EA, CCL8 was associated with decreased OS. Expression of CCL5, -7, -8, -17, -20 and -25 was highest in TNBC. Expression of CCL11 and CCL22 was associated with HER2. CCL7, -8, -17, -20 and -25 were elevated in AAs. In conclusion, our analysis suggests significant association of CC-chemokines in BrCa progression, OS and disparate disease outcome in AA compared to EA patients.
Collapse
|
41
|
Xu Y, Dong X, Qi P, Ye Y, Shen W, Leng L, Wang L, Li X, Luo X, Chen Y, Sun P, Xiang R, Li N. Sox2 Communicates with Tregs Through CCL1 to Promote the Stemness Property of Breast Cancer Cells. Stem Cells 2018; 35:2351-2365. [PMID: 29044882 DOI: 10.1002/stem.2720] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 07/10/2017] [Indexed: 01/10/2023]
Abstract
As an important component of the tumor microenvironment, CD4+ CD25+ Tregs reduce antitumor immunity, promote angiogenesis and metastasis in breast cancer. However, their function in regulating the "stemness" of tumor cells and the communication between Tregs and cancer stem cells (CSCs) remain elusive. Here, we disclose that the primarily cultured Tregs isolated from breast-tumor-bearing Foxp3-EGFP mouse upregulate the stemness property of breast cancer cells. Tregs increased the side-population and the Aldehyde dehydrogenase-bright population of mouse breast cancer cells, promoted their sphere formation in a paracrine manner, and enhanced the expression of stemness genes, such as Sox2 and so forth. In addition, Tregs increased tumorigenesis, metastasis, and chemoresistance of breast cancer cells. Furthermore, Sox2-overexpression tumor cells activated NF-κB-CCL1 signaling to recruit Tregs through reducing the binding of H3K27Me3 on promoter regions of p65 and Ccl1. These findings reveal the functional interaction between Tregs and CSCs and indicate that targeting on the communication between them is a promising strategy in breast cancer therapy. Stem Cells 2017;35:2351-2365.
Collapse
Affiliation(s)
- Yingxi Xu
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China.,State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, People's Republic of China
| | - Xiaoli Dong
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Pingping Qi
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Yujie Ye
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Wenzhi Shen
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Liang Leng
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Lina Wang
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China.,State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, People's Republic of China
| | - Xuefei Li
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Medical Technology, Beijing Health Vocational College, Beijing, People's Republic of China
| | - Xiaohe Luo
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Yanan Chen
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Peiqing Sun
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Rong Xiang
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China.,Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University, Tianjin, People's Republic of China.,Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin, People's Republic of China
| | - Na Li
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University, Tianjin, People's Republic of China.,Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin, People's Republic of China.,Department of Pathophysiology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| |
Collapse
|
42
|
Prasmickaite L, Tenstad EM, Pettersen S, Jabeen S, Egeland EV, Nord S, Pandya A, Haugen MH, Kristensen VN, Børresen-Dale AL, Engebråten O, Maelandsmo GM. Basal-like breast cancer engages tumor-supportive macrophages via secreted factors induced by extracellular S100A4. Mol Oncol 2018; 12:1540-1558. [PMID: 29741811 PMCID: PMC6120223 DOI: 10.1002/1878-0261.12319] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/22/2018] [Accepted: 04/09/2018] [Indexed: 12/14/2022] Open
Abstract
The tumor microenvironment (TME) may influence both cancer progression and therapeutic response. In breast cancer, particularly in the aggressive triple‐negative/basal‐like subgroup, patient outcome is strongly associated with the tumor's inflammatory profile. Tumor‐associated macrophages (TAMs) are among the most abundant immune cells in the TME, shown to be linked to poor prognosis and therapeutic resistance. In this study, we investigated the effect of the metastasis‐ and inflammation‐associated microenvironmental factor S100A4 on breast cancer cells (BCCs) of different subtypes and explored their further interactions with myeloid cells. We demonstrated that extracellular S100A4 activates BCCs, particularly the basal‐like subtype, to elevate secretion of pro‐inflammatory cytokines. The secreted factors promoted conversion of monocytes to TAM‐like cells that exhibited protumorigenic activities: stimulated epithelial–mesenchymal transition, proliferation, chemoresistance, and motility in cancer cells. In conclusion, we have shown that extracellular S100A4 instigates a tumor‐supportive microenvironment, involving a network of cytokines and TAM‐like cells, which was particularly characteristic for basal‐like BCCs and potentiated their aggressive properties. The S100A4–BCC–TAM interaction cascade could be an important contributor to the aggressive behavior of this subtype and should be further explored for therapeutic targeting.
Collapse
Affiliation(s)
- Lina Prasmickaite
- Department of Tumor Biology, Institute of Cancer Research, Oslo University Hospital, Norway
| | - Ellen M Tenstad
- Department of Tumor Biology, Institute of Cancer Research, Oslo University Hospital, Norway
| | - Solveig Pettersen
- Department of Tumor Biology, Institute of Cancer Research, Oslo University Hospital, Norway
| | - Shakila Jabeen
- Department of Clinical Molecular Biology (EpiGen), Akershus University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Eivind V Egeland
- Department of Tumor Biology, Institute of Cancer Research, Oslo University Hospital, Norway
| | - Silje Nord
- Department of Cancer Genetics, Institute of Cancer Research, Oslo University Hospital, Norway
| | - Abhilash Pandya
- Department of Tumor Biology, Institute of Cancer Research, Oslo University Hospital, Norway
| | - Mads H Haugen
- Department of Tumor Biology, Institute of Cancer Research, Oslo University Hospital, Norway
| | - Vessela N Kristensen
- Department of Clinical Molecular Biology (EpiGen), Akershus University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway.,Department of Cancer Genetics, Institute of Cancer Research, Oslo University Hospital, Norway
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute of Cancer Research, Oslo University Hospital, Norway
| | | | - Olav Engebråten
- Department of Tumor Biology, Institute of Cancer Research, Oslo University Hospital, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway.,Department of Oncology, Oslo University Hospital, Norway
| | - Gunhild M Maelandsmo
- Department of Tumor Biology, Institute of Cancer Research, Oslo University Hospital, Norway.,Department of Medical Biology, Faculty of Health Sciences, UiT/The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
43
|
Kennedy E, Hokmabadi M, Dong Z, McKelvey K, Nelson EM, Timp G. Method for Dynamically Detecting Secretions from Single Cells Using a Nanopore. NANO LETTERS 2018; 18:4263-4272. [PMID: 29870666 DOI: 10.1021/acs.nanolett.8b01257] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Secreted proteins mediate cell-to-cell communications. Thus, eavesdropping on the secretome could reveal the cellular phenotype, but it is challenging to detect the proteins because they are secreted only in minute amounts and then diluted in blood plasma or contaminated by cell culture medium or the lysate. In this pilot study, it is demonstrated that secretions from single cancer cells can be detected and dynamically analyzed through measurements of blockades in the electrolytic current due to single molecules translocating through a nanopore in a thin inorganic membrane. It is established that the distribution of blockades can be used to differentiate three different cancer cell lines (U937, MDA-MB-231, and MCF-7) in real time and quickly (<20 s). Importantly, the distinctive blockades associated with the chemokine CCL5, a prognostic factor for disease progression in breast cancer, along with other low-mass biomarkers of breast cancer (PI3, TIMP1, and MMP1) were identified in the context of the secretome of these three cell types, tracked with time, and used to provide information on the cellular phenotype.
Collapse
|
44
|
Ma G, Huang H, Li M, Li L, Kong P, Zhu Y, Xia T, Wang S. Plasma CCL5 promotes EMT-medicated epirubicin-resistance in locally advanced breast cancer. Cancer Biomark 2018; 22:405-415. [PMID: 29758926 DOI: 10.3233/cbm-170986] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Neoadjuvant chemotherapy (NCT) is the standard treatment for locally advanced breast cancer (LABC). Pathological complete response (pCR) is commonly used as a valid predictor of NCT long-term outcomes. Blood-based tumor biomarkers have the potential to predict response to NCT at early stage non-invasively. We believed plasma CCL5 could be a potential marker to predict NCT of LABC. Its efficiency and possible mechanism was studied in this work. Human Cytokine Antibody Microarray was applied to screen different cytokine concentration in plasma between low histological regression (Low-R) and high histological regression (High-R) patients. LABC patients were divided into two groups according to pathological reactivity. The concentration of plasma CCL5 in different groups was determined by ELISA analysis. CCK8 assay was performed to analyze epirubicin susceptibility of breast cancer cells. Transwell assay was performed to determine the effect of CCL5 on breast cancer cells' migration and invasion. qRT-PCR and western blot were used to verify the EMT (epithelial-mesenchymal transition) markers in CCL5-treated and epirubicin-treated breast cancer cells. The concentration of plasma CCL5 of Low-R group was higher than High-R group before NCT. The plasma levels of CCL5 were significantly reduced after NCT in the group of high histological regression (High-R). Epirubicin susceptibility decreased in the breast cancer cells treated by recombinant CCL5. Migration and invasion were significantly enhanced in breast cancer cells treated by recombinant CCL5. E-cadherin expression was decreased whereas vimentin increased significantly in CCL5-treated breast cancer cells. The phosphorylation of ezrin in Y-567 and its downstream protein cortactin increased significantly in CCL5-treated breast cancer cells. Plasma CCL5 level could be a promised candidate to predict chemotherapy response of breast cancer. Plasma CCL5 plays an important role in EMT process of breast cancer.
Collapse
Affiliation(s)
- Ge Ma
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 210029 Nanjing, Jiangsu, China.,Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Huaxing Huang
- The First Clinical Medical College, Nanjing Medical University, 210029 Nanjing, Jiangsu, China.,Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Minghui Li
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 210029 Nanjing, Jiangsu, China.,Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Li Li
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Peng Kong
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Yichao Zhu
- Department of Physiology, Nanjing Medical University, Nanjing 211166, Jiangsu, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Tiansong Xia
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Shui Wang
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| |
Collapse
|
45
|
Newcomb B, Rhein C, Mileva I, Ahmad R, Clarke CJ, Snider J, Obeid LM, Hannun YA. Identification of an acid sphingomyelinase ceramide kinase pathway in the regulation of the chemokine CCL5. J Lipid Res 2018; 59:1219-1229. [PMID: 29724781 DOI: 10.1194/jlr.m084202] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/25/2018] [Indexed: 02/01/2023] Open
Abstract
Acid sphingomyelinase (ASM) hydrolyzes sphingomyelin to produce the biologically active lipid ceramide. Previous studies have implicated ASM in the induction of the chemokine CCL5 in response to TNF-α however, the lipid mediator of this effect was not established. In the present study, we identified a novel pathway connecting ASM and ceramide kinase (CERK). The results show that TNF-α induces the formation of ceramide 1-phosphate (C-1-P) in a CERK-dependent manner. Silencing of CERK blocks CCL5 production in response to TNF-α. Interestingly, cells lacking ASM have decreased C-1-P production following TNF-α treatment, suggesting that ASM may be acting upstream of CERK. Functionally, ASM and CERK induce a highly concordant program of cytokine production and both are required for migration of breast cancer cells. Taken together, these data suggest ASM can produce ceramide which is then converted to C-1-P by CERK, and that C-1-P is required for production of CCL5 and several cytokines and chemokines, with roles in cell migration. These results highlight the diversity in action of ASM through more than one bioactive sphingolipid.
Collapse
Affiliation(s)
- Benjamin Newcomb
- Stony Brook Cancer Center Stony Brook University, Stony Brook, NY 11794
| | - Cosima Rhein
- Stony Brook Cancer Center Stony Brook University, Stony Brook, NY 11794.,Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Izolda Mileva
- Stony Brook Cancer Center Stony Brook University, Stony Brook, NY 11794
| | - Rasheed Ahmad
- Immunology and Innovative Cell Therapy Unit, Dasman Diabetes Institute, Kuwait City, Kuwait
| | | | - Justin Snider
- Stony Brook Cancer Center Stony Brook University, Stony Brook, NY 11794
| | - Lina M Obeid
- Stony Brook Cancer Center Stony Brook University, Stony Brook, NY 11794.,Department of Medicine, Stony Brook University, Stony Brook, NY 11794
| | - Yusuf A Hannun
- Stony Brook Cancer Center Stony Brook University, Stony Brook, NY 11794 .,Department of Medicine, Stony Brook University, Stony Brook, NY 11794
| |
Collapse
|
46
|
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a key regulator of numerous physiological functions, including the immune response. As pathogens elicit an acute phase response with concerted activation of STAT3, they are confronted with two evolutionary options: either curtail it or employ it. This has important consequences for the host, since abnormal STAT3 function is associated with cancer development and other diseases. This review provides a comprehensive outline of how human viruses cope with STAT3-mediated inflammation and how this affects the host. Finally, we discuss STAT3 as a potential target for antiviral therapy.
Collapse
Affiliation(s)
- Armando Andres Roca Suarez
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Nicolaas Van Renne
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Thomas F. Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Pôle Hépato-digestif, Institut Hospitalo-universitaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Joachim Lupberger
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- * E-mail:
| |
Collapse
|
47
|
Mandal PK, Biswas S, Mandal G, Purohit S, Gupta A, Majumdar (Giri) A, Roy Chowdhury S, Bhattacharyya A. CCL2 conditionally determines CCL22-dependent Th2-accumulation during TGF-β-induced breast cancer progression. Immunobiology 2018; 223:151-161. [DOI: 10.1016/j.imbio.2017.10.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 10/14/2017] [Indexed: 12/12/2022]
|
48
|
CCR5/CCL5 axis interaction promotes migratory and invasiveness of pancreatic cancer cells. Sci Rep 2018; 8:1323. [PMID: 29358632 PMCID: PMC5778036 DOI: 10.1038/s41598-018-19643-0] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers and remains a major challenge due to its invasive and metastatic nature. Increased levels of CCR5 and CCL5 have established indicators for disease status in various cancers, including PC. However, their role in invasion and metastasis of PC is not known. Here we conducted immunohistochemistry of PC tissues and found elevated epithelial staining for CCR5 and CCL5 in metastatic PC tissues compared to non-neoplastic. In vitro experiments, such as flow cytometry, immunofluorescence and western blotting with human PC cell lines (AsPc-1, BxPc-3 and MIA PaCa-2), showed higher expression levels of CCR5. The CCL5 activation of PC cells expressing CCR5 increased their invasive potential, while treatment with CCR5 inhibitor maraviroc inhibited the CCL5 activation. CCL5 induced proliferation of PC cells was mediated through F-actin polymerization, while there was marked reduction when the cells were treated with maraviroc. The direct interaction of CCR5 with CCL5 was verified using a calcium mobilization assay. Taken together, our results demonstrate that CCR5 and CCL5 are potential markers for metastatic PC cancer, and their interaction leads to the increased PC cell invasion. Thus, blocking CCR5/CCL5 axis might prove beneficial to prevent metastasis and provide a more therapeutic strategy to control PC progression.
Collapse
|
49
|
Allen H, Shraga-Heled N, Blumenfeld M, Dego-Ashto T, Fuchs-Telem D, Gilert A, Aberman Z, Ofir R. Human Placental-Derived Adherent Stromal Cells Co-Induced with TNF-α and IFN-γ Inhibit Triple-Negative Breast Cancer in Nude Mouse Xenograft Models. Sci Rep 2018; 8:670. [PMID: 29330447 PMCID: PMC5766494 DOI: 10.1038/s41598-017-18428-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022] Open
Abstract
Culturing 3D-expanded human placental-derived adherent stromal cells (ASCs) in the presence of tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) transiently upregulated the secretion of numerous anti-proliferative, anti-angiogenic and pro-inflammatory cytokines. In a 3D-spheroid screening assay, conditioned medium from these induced-ASCs inhibited proliferation of cancer cell lines, including triple-negative breast cancer (TNBC) lines. In vitro co-culture studies of induced-ASCs with MDA-MB-231 human breast carcinoma cells, a model representing TNBC, supports a mechanism involving immunomodulation and angiogenesis inhibition. In vivo studies in nude mice showed that intramuscular administration of induced-ASCs halted MDA-MB-231 cell proliferation, and inhibited tumor progression and vascularization. Thirty percent of treated mice experienced complete tumor remission. Murine serum concentrations of the tumor-supporting cytokines Interleukin-6 (IL-6), Vascular endothelial growth factor (VEGF) and Granulocyte-colony stimulating factor (G-CSF) were lowered to naïve levels. A somatic mutation analysis identified numerous genes which could be screened in patients to increase a positive therapeutic outcome. Taken together, these results show that targeted changes in the secretion profile of ASCs may improve their therapeutic potential.
Collapse
|
50
|
Gao D, Cazares LH, Fish EN. CCL5-CCR5 interactions modulate metabolic events during tumor onset to promote tumorigenesis. BMC Cancer 2017; 17:834. [PMID: 29216863 PMCID: PMC5721608 DOI: 10.1186/s12885-017-3817-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/22/2017] [Indexed: 01/16/2023] Open
Abstract
Background In earlier studies we have shown that CCL5 activation of CCR5 induces the proliferation and survival of breast cancer cells in a mechanistic target of rapamycin (mTOR)-dependent manner and that this is in part due to CCR5-mediated increases in glycolytic metabolism. Methods Using the MDA-MB-231 triple negative human breast cancer cell line and mouse mammary tumor virus – polyomavirus middle T-antigen (MMTV-PyMT) mouse primary breast cancer cells, we conducted in vivo tumor transplant experiments to examine the effects of CCL5-CCR5 interactions in the context of regulating tumor metabolism. Additionally, we employed Matrix-Assisted Laser Desorption/Ionization Fourier Transform Ion Cyclotron Resonance Mass Spectrometry imaging (MALDI-FTICR-MSI) to evaluate tumor utilization of cellular metabolites. Results We provide evidence that, in the absence of CCR5, the early events associated with rapid tumor growth in the MMTV-PyMT mouse model of spontaneous breast cancer development, are diminished, as demonstrated by a delay in tumor onset. In tumor transplant studies into immunocompromised mice we identify a direct correlation between reduced tumor proliferation and decreased metabolic activity, specifically associated with tumor expression of CCR5. The reduction in tumorigenesis is accompanied by decreases in glucose uptake, glucose transporter-1 (GLUT-1) cell surface expression, intracellular ATP and lactate levels, as well as reduced CCL5 production. Using MALDI-FTICR-MS, we show that the rapid early tumor growth of CCR5+/+ triple negative breast cancer cells in vivo is attributable to increased levels of glycolytic intermediates required for anabolic processes, in contrast to the slower growth rate of their corresponding CCR5−/− cells, that exhibit reduced glycolytic metabolism. Conclusions These findings suggest that CCL5-CCR5 interactions in the tumor microenvironment modulate metabolic events during tumor onset to promote tumorigenesis. Electronic supplementary material The online version of this article (10.1186/s12885-017-3817-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Darrin Gao
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.,Department of Immunology, University of Toronto, Toronto, Canada
| | - Lisa H Cazares
- Molecular and Translational Sciences Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, USA
| | - Eleanor N Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada. .,Department of Immunology, University of Toronto, Toronto, Canada.
| |
Collapse
|