1
|
Kim G, Kim EY, Lee H, Shin SH, Lee SH, Sohn KY, Kim JW, Lee JS. 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol treatment inhibits abnormal tumor growth by regulating neutrophil infiltration in a non-small cell lung carcinoma mouse model. Biomed Pharmacother 2024; 178:117269. [PMID: 39137654 DOI: 10.1016/j.biopha.2024.117269] [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: 05/07/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Excessive neutrophil infiltration into the tumor microenvironment (TME) is an important factor that contributes to tumor overgrowth and limited immunotherapy efficacy. Neutrophils activate various receptors involved in tumor progression, while suppressing the infiltration and activity of cytotoxic T cells and creating optimal conditions for tumor growth. Therefore, the appropriate control of neutrophil infiltration is an effective strategy for tumor treatment. In the present study, 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) inhibited tumor overgrowth by suppressing excessive neutrophil infiltration, resulting in >74.97 % reduction in tumor size in a Lewis lung carcinoma (LLC-1) mouse model. All subjects in the positive control group died during the 90-day survival period, whereas only four subjects in the PLAG treatment group survived. PLAG had a significantly higher tumor growth inhibitory effect and survival rate than other neutrophil infiltration-targeting inhibitors (e.g., Navarixin, lymphocyte antigen 6 complex locus G6D antibody [aLy6G]). The ability of PLAG to regulate neutrophil infiltration and inhibit tumor growth depends on thioredoxin-interacting protein (TXNIP). In tumors lacking TXNIP expression, PLAG failed to control neutrophil infiltration and infiltration-related factor release, and the inhibitory effect of PLAG on tumor growth was reduced. PLAG-mediated inhibition of neutrophil infiltration enhances the efficacy of immune checkpoint inhibitors (ICIs), increasing the antitumor efficacy and survival rate by 30 %. In conclusion, PLAG could be a novel alternative to anti-tumor drugs that effectively targets excessive neutrophil infiltration into cancer tissues.
Collapse
Affiliation(s)
- Guentae Kim
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea; Biotoxtech, 53 Yeongudanji-ro, Ochang-eup, Cheongju-si, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea
| | - Jae Sam Lee
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea.
| |
Collapse
|
2
|
Roberts BK, Li DI, Somerville C, Matta B, Jha V, Steinke A, Brune Z, Blanc L, Soffer SZ, Barnes BJ. IRF5 suppresses metastasis through the regulation of tumor-derived extracellular vesicles and pre-metastatic niche formation. Sci Rep 2024; 14:15557. [PMID: 38969706 PMCID: PMC11226449 DOI: 10.1038/s41598-024-66168-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024] Open
Abstract
Metastasis is driven by extensive cooperation between a tumor and its microenvironment, resulting in the adaptation of molecular mechanisms that evade the immune system and enable pre-metastatic niche (PMN) formation. Little is known of the tumor-intrinsic factors that regulate these mechanisms. Here we show that expression of the transcription factor interferon regulatory factor 5 (IRF5) in osteosarcoma (OS) and breast carcinoma (BC) clinically correlates with prolonged survival and decreased secretion of tumor-derived extracellular vesicles (t-dEVs). Conversely, loss of intra-tumoral IRF5 establishes a PMN that supports metastasis. Mechanistically, IRF5-positive tumor cells retain IRF5 transcripts within t-dEVs that contribute to altered composition, secretion, and trafficking of t-dEVs to sites of metastasis. Upon whole-body pre-conditioning with t-dEVs from IRF5-high or -low OS and BC cells, we found increased lung metastatic colonization that replicated findings from orthotopically implanted cancer cells. Collectively, our findings uncover a new role for IRF5 in cancer metastasis through its regulation of t-dEV programming of the PMN.
Collapse
Affiliation(s)
- Bailey K Roberts
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Dan Iris Li
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Carter Somerville
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Bharati Matta
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Vaishali Jha
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | | | - Zarina Brune
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Lionel Blanc
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Departments of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, 11549, USA
| | - Samuel Z Soffer
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Department of Pediatric Surgery, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, 11549, USA
| | - Betsy J Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
- Departments of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, 11549, USA.
| |
Collapse
|
3
|
Qiao Q, Hu S, Wang X. The regulatory roles and clinical significance of glycolysis in tumor. Cancer Commun (Lond) 2024; 44:761-786. [PMID: 38851859 PMCID: PMC11260772 DOI: 10.1002/cac2.12549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/05/2024] [Accepted: 05/12/2024] [Indexed: 06/10/2024] Open
Abstract
Metabolic reprogramming has been demonstrated to have a significant impact on the biological behaviors of tumor cells, among which glycolysis is an important form. Recent research has revealed that the heightened glycolysis levels, the abnormal expression of glycolytic enzymes, and the accumulation of glycolytic products could regulate the growth, proliferation, invasion, and metastasis of tumor cells and provide a favorable microenvironment for tumor development and progression. Based on the distinctive glycolytic characteristics of tumor cells, novel imaging tests have been developed to evaluate tumor proliferation and metastasis. In addition, glycolytic enzymes have been found to serve as promising biomarkers in tumor, which could provide assistance in the early diagnosis and prognostic assessment of tumor patients. Numerous glycolytic enzymes have been identified as potential therapeutic targets for tumor treatment, and various small molecule inhibitors targeting glycolytic enzymes have been developed to inhibit tumor development and some of them are already applied in the clinic. In this review, we systematically summarized recent advances of the regulatory roles of glycolysis in tumor progression and highlighted the potential clinical significance of glycolytic enzymes and products as novel biomarkers and therapeutic targets in tumor treatment.
Collapse
Affiliation(s)
- Qiqi Qiao
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
| | - Shunfeng Hu
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
| | - Xin Wang
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
- Taishan Scholars Program of Shandong ProvinceJinanShandongP. R. China
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongP. R. China
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuP. R. China
| |
Collapse
|
4
|
Chen Y, Ouyang D, Wang Y, Pan Q, Zhao J, Chen H, Yang X, Tang Y, Wang Q, Li Y, He J, You JQ, Li Y, Xu C, Ren Y, Xie S, Li S, Lian J, Weng D, Xiang T, Xia JC. EBV promotes TCR-T-cell therapy resistance by inducing CD163+M2 macrophage polarization and MMP9 secretion. J Immunother Cancer 2024; 12:e008375. [PMID: 38886114 PMCID: PMC11184188 DOI: 10.1136/jitc-2023-008375] [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] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV) is a double-stranded DNA oncogenic virus. Several types of solid tumors, such as nasopharyngeal carcinoma, EBV-associated gastric carcinoma, and lymphoepithelioma-like carcinoma of the lung, have been linked to EBV infection. Currently, several TCR-T-cell therapies for EBV-associated tumors are in clinical trials, but due to the suppressive immune microenvironment of solid tumors, the clinical application of TCR-T-cell therapy for EBV-associated solid tumors is limited. Figuring out the mechanism by which EBV participates in the formation of the tumor immunosuppressive microenvironment will help T cells or TCR-T cells break through the limitation and exert stronger antitumor potential. METHODS Flow cytometry was used for analyzing macrophage differentiation phenotypes induced by EBV-infected and EBV-uninfected tumors, as well as the function of T cells co-cultured with these macrophages. Xenograft model in mice was used to explore the effects of M2 macrophages, TCR-T cells, and matrix metalloprotein 9 (MMP9) inhibitors on the growth of EBV-infected tumors. RESULTS EBV-positive tumors exhibited an exhaustion profile of T cells, despite the presence of a large T-cell infiltration. EBV-infected tumors recruited a large number of mononuclear macrophages with CCL5 and induced CD163+M2 macrophages polarization through the secretion of CSF1 and the promotion of autocrine IL10 production by mononuclear macrophages. Massive secretion of MMP9 by this group of CD163+M2 macrophages induced by EBV infection was an important factor contributing to T-cell exhaustion and TCR-T-cell therapy resistance in EBV-positive tumors, and the use of MMP9 inhibitors improved the function of T cells cocultured with M2 macrophages. Finally, the combination of an MMP9 inhibitor with TCR-T cells targeting EBV-positive tumors significantly inhibited the growth of xenografts in mice. CONCLUSIONS MMP9 inhibitors improve TCR-T cell function suppressed by EBV-induced M2 macrophages. TCR-T-cell therapy combined with MMP9 inhibitors was an effective therapeutic strategy for EBV-positive solid tumors.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Dijun Ouyang
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yan Wang
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Qiuzhong Pan
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jingjing Zhao
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Hao Chen
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xinyi Yang
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yan Tang
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Qijing Wang
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yongqiang Li
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jia He
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jin-Qi You
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yingzi Li
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Chi Xu
- Knowcell Biotechnology Co., Ltd, Shenzhen, China
| | - Yan Ren
- Knowcell Biotechnology Co., Ltd, Shenzhen, China
| | - Sisi Xie
- Knowcell Biotechnology Co., Ltd, Shenzhen, China
| | - Song Li
- TCRCure Biological Technology Co., Ltd, Guangzhou, China
| | - Jiamin Lian
- TCRCure Biological Technology Co., Ltd, Guangzhou, China
| | - Desheng Weng
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Tong Xiang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jian-Chuan Xia
- Department of Biotherapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Tanaka Y, Ito T, Murata M, Tanegashima K, Kaku-Ito Y, Nakahara T. NECTIN4-targeted antibody-drug conjugate is a potential therapeutic option for extramammary Paget disease. Exp Dermatol 2024; 33:e15049. [PMID: 38509717 DOI: 10.1111/exd.15049] [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: 01/04/2024] [Revised: 02/15/2024] [Accepted: 02/24/2024] [Indexed: 03/22/2024]
Abstract
Extramammary Paget disease (EMPD) is a rare skin cancer mainly found in areas rich in apocrine sweat glands. Since the effective treatments for advanced and/or metastasized EMPD are limited, there is an urgent need to develop novel therapeutic approaches. Nectin cell adhesion molecule 4 (NECTIN4) is highly expressed in cancers and considered to be a promising therapeutic target. NECTIN4 is also expressed in EMPD, but its role and the efficacy of NECTIN4-targeted therapy in EMPD remain unclear. This study investigated the potential of NECTIN4 as a novel therapeutic target for EMPD. NECTIN4 expression was immunohistochemically analysed in EMPD patients' primary (118 samples) and metastatic (21 samples) lesions. Using an EMPD cell line, KS-EMPD-1, the effects of NECTIN4 inhibition on cell proliferation and migration were investigated. NECTIN4 was expressed in primary and metastatic EMPD lesions, and the H-score of NECTIN4 staining was significantly higher in metastatic lesions than in primary ones. Knockdown of NECTIN4 significantly inhibited cell proliferation and affected cell migration. The cytotoxic effects of NECTIN4-targeted antibody-drug conjugate (ADC) were further evaluated, revealing a significant decrease in EMPD cell viability. In conclusion, NECTIN4 is a potential therapeutic target and NECTIN4-targeted ADC is promising as a therapeutic option for EMPD.
Collapse
Affiliation(s)
- Yuka Tanaka
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Maho Murata
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keiko Tanegashima
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yumiko Kaku-Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
6
|
Xu W, Goreczny GJ, Forsythe I, Brennan G, Stowell T, Brock K, Capella B, Turner CE. Hic-5 regulates extracellular matrix-associated gene expression and cytokine secretion in cancer associated fibroblasts. Exp Cell Res 2024; 435:113930. [PMID: 38237846 PMCID: PMC10923124 DOI: 10.1016/j.yexcr.2024.113930] [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: 09/21/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
The focal adhesion protein, Hic-5 plays a key role in promoting extracellular matrix deposition and remodeling by cancer associated fibroblasts within the tumor stroma to promote breast tumor cell invasion. However, whether stromal matrix gene expression is regulated by Hic-5 is still unknown. Utilizing a constitutive Hic-5 knockout, Mouse Mammary Tumor Virus-Polyoma Middle T-Antigen spontaneous breast tumor mouse model, bulk RNAseq analysis was performed on cancer associated fibroblasts isolated from Hic-5 knockout mammary tumors. Functional network analysis highlighted a key role for Hic-5 in extracellular matrix organization, with both structural matrix genes, as well as matrix remodeling genes being differentially expressed in relation to Hic-5 expression. The subcellular distribution of the MRTF-A transcription factor and expression of a subset of MRTF-A responsive genes was also impacted by Hic-5 expression. Additionally, cytokine array analysis of conditioned media from the Hic-5 and Hic-5 knockout cancer associated fibroblasts revealed that Hic-5 is important for the secretion of several key factors that are associated with matrix remodeling, angiogenesis and immune evasion. Together, these data provide further evidence of a central role for Hic-5 expression in cancer associated fibroblasts in regulating the composition and organization of the tumor stroma microenvironment to promote breast tumor progression.
Collapse
Affiliation(s)
- Weiyi Xu
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Gregory J Goreczny
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Jnana Therapeutics, Boston, MA, USA
| | - Ian Forsythe
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Zymo Research Corp, Huntington Beach, CA, USA
| | - Grant Brennan
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Theresa Stowell
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Katia Brock
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Benjamin Capella
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Christopher E Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
7
|
Wei C. The multifaceted roles of matrix metalloproteinases in lung cancer. Front Oncol 2023; 13:1195426. [PMID: 37766868 PMCID: PMC10520958 DOI: 10.3389/fonc.2023.1195426] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/08/2023] [Indexed: 09/29/2023] Open
Abstract
Background Though the matrix metalloproteinases (MMPs) are widely investigated in lung cancer (LC), however, almost no review systematically clarify their multi-faced roles in LC. Methods We investigated the expression of MMPs and their effects on survival of patients with LC, the resistance mechanisms of MMPs in anti-tumor therapy, the regulatory networks of MMPs involved, the function of MMPs inducing CSCLs, MMPs-related tumor immunity, and effects of MMP polymorphisms on risk of LC. Results High expression of MMPs was mainly related to poor survival, high clinical stages and cancer metastasis. Role of MMPs in LC are multi-faced. MMPs are involved in drug resistance, induced CSCLs, participated in tumor immunity. Besides, MMPs polymorphisms may increase risk of LC. Conclusions MMPs might be promising targets to restore the anti-tumor immune response and enhance the killing function of nature immune cells in LC.
Collapse
Affiliation(s)
- Cui Wei
- Department of Emergency, The Third Hospital of Changsha, Changsha, China
| |
Collapse
|
8
|
Zhang S, Regan K, Najera J, Grinstaff MW, Datta M, Nia HT. The peritumor microenvironment: physics and immunity. Trends Cancer 2023; 9:609-623. [PMID: 37156677 PMCID: PMC10523902 DOI: 10.1016/j.trecan.2023.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 05/10/2023]
Abstract
Cancer initiation and progression drastically alter the microenvironment at the interface between healthy and malignant tissue. This site, termed the peritumor, bears unique physical and immune attributes that together further promote tumor progression through interconnected mechanical signaling and immune activity. In this review, we describe the distinct physical features of the peritumoral microenvironment and link their relationship to immune responses. The peritumor is a region rich in biomarkers and therapeutic targets and thus is a key focus for future cancer research as well as clinical outlooks, particularly to understand and overcome novel mechanisms of immunotherapy resistance.
Collapse
Affiliation(s)
- Sue Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Kathryn Regan
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Julian Najera
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA, USA; Department of Chemistry, Boston University, Boston, MA, USA
| | - Meenal Datta
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA.
| | - Hadi T Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| |
Collapse
|
9
|
Rashid ZA, Bardaweel SK. Novel Matrix Metalloproteinase-9 (MMP-9) Inhibitors in Cancer Treatment. Int J Mol Sci 2023; 24:12133. [PMID: 37569509 PMCID: PMC10418771 DOI: 10.3390/ijms241512133] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Matrix metalloproteinases (MMPs) belong to a family of zinc-dependent proteolytic metalloenzymes. MMP-9, a member of the gelatinase B family, is characterized as one of the most intricate MMPs. The crucial involvement of MMP-9 in extracellular matrix (ECM) remodeling underscores its significant correlation with each stage of cancer pathogenesis and progression. The design and synthesis of MMP-9 inhibitors is a potentially attractive research area. Unfortunately, to date, there is no effective MMP-9 inhibitor that passes the clinical trials and is approved by the FDA. This review primarily focuses on exploring the diverse strategies employed in the design and advancement of MMP-9 inhibitors, along with their anticancer effects and selectivity. To illuminate the essential structural characteristics necessary for the future design of novel MMP-9 inhibitors, the current narrative review highlights several recently discovered MMP-9 inhibitors exhibiting notable selectivity and potency.
Collapse
Affiliation(s)
| | - Sanaa K. Bardaweel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
| |
Collapse
|
10
|
He L, Kang Q, Chan KI, Zhang Y, Zhong Z, Tan W. The immunomodulatory role of matrix metalloproteinases in colitis-associated cancer. Front Immunol 2023; 13:1093990. [PMID: 36776395 PMCID: PMC9910179 DOI: 10.3389/fimmu.2022.1093990] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/07/2022] [Indexed: 01/22/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are an important class of enzymes in the body that function through the extracellular matrix (ECM). They are involved in diverse pathophysiological processes, such as tumor invasion and metastasis, cardiovascular diseases, arthritis, periodontal disease, osteogenesis imperfecta, and diseases of the central nervous system. MMPs participate in the occurrence and development of numerous cancers and are closely related to immunity. In the present study, we review the immunomodulatory role of MMPs in colitis-associated cancer (CAC) and discuss relevant clinical applications. We analyze more than 300 pharmacological studies retrieved from PubMed and the Web of Science, related to MMPs, cancer, colitis, CAC, and immunomodulation. Key MMPs that interfere with pathological processes in CAC such as MMP-2, MMP-3, MMP-7, MMP-9, MMP-10, MMP-12, and MMP-13, as well as their corresponding mechanisms are elaborated. MMPs are involved in cell proliferation, cell differentiation, angiogenesis, ECM remodeling, and the inflammatory response in CAC. They also affect the immune system by modulating differentiation and immune activity of immune cells, recruitment of macrophages, and recruitment of neutrophils. Herein we describe the immunomodulatory role of MMPs in CAC to facilitate treatment of this special type of colon cancer, which is preceded by detectable inflammatory bowel disease in clinical populations.
Collapse
Affiliation(s)
- Luying He
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Qianming Kang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Ka Iong Chan
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Yang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China,*Correspondence: Zhangfeng Zhong, ; Wen Tan,
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou, China,*Correspondence: Zhangfeng Zhong, ; Wen Tan,
| |
Collapse
|
11
|
Kwon MJ. Matrix metalloproteinases as therapeutic targets in breast cancer. Front Oncol 2023; 12:1108695. [PMID: 36741729 PMCID: PMC9897057 DOI: 10.3389/fonc.2022.1108695] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/28/2022] [Indexed: 01/22/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are the most prominent proteinases involved in tumorigenesis. They were initially recognized to promote tumor progression by remodeling the extracellular matrix through their proteolytic activity. However, accumulating evidence has revealed that some MMPs have protective roles in cancer progression, and the same MMP can exert opposing roles depending on the cell type in which it is expressed or the stage of cancer. Moreover, studies have shown that MMPs are involved in cancer progression through their roles in other biological processes such as cell signaling and immune regulation, independent of their catalytic activity. Despite the prognostic significance of tumoral or stromal expression of MMPs in breast cancer, their roles and molecular mechanisms in breast cancer progression remain unclear. As the failures of early clinical trials with broad-spectrum MMP inhibitors were mainly due to a lack of drug specificity, substantial efforts have been made to develop highly selective MMP inhibitors. Some recently developed MMP inhibitory monoclonal antibodies demonstrated promising anti-tumor effects in preclinical models of breast cancer. Importantly, anti-tumor effects of these antibodies were associated with the modulation of tumor immune microenvironment, suggesting that the use of MMP inhibitors in combination with immunotherapy can improve the efficacy of immunotherapy in HER2-positive or triple-negative breast cancer. In this review, the current understanding of the roles of tumoral or stromal MMPs in breast cancer is summarized, and recent advances in the development of highly selective MMP inhibitors are discussed.
Collapse
Affiliation(s)
- Mi Jeong Kwon
- Vessel-Organ Interaction Research Center (MRC), College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea,BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea,*Correspondence: Mi Jeong Kwon,
| |
Collapse
|
12
|
li M, Yan T, Cai Y, Wei Y, Xie Q. Expression of matrix metalloproteinases and their association with clinical characteristics of solid tumors. Gene X 2023; 850:146927. [DOI: 10.1016/j.gene.2022.146927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022] Open
|
13
|
Ye L, Jiang Y, Zhang M. Crosstalk between glucose metabolism, lactate production and immune response modulation. Cytokine Growth Factor Rev 2022; 68:81-92. [PMID: 36376165 DOI: 10.1016/j.cytogfr.2022.11.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
Abstract
Metabolites of glycolytic metabolism have been identified as signaling molecules and regulators of gene expression, in addition to their basic function as major energy and biosynthetic source. Immune cells reprogram metabolic pathways to cater to energy and biosynthesis demands upon activation. Most lymphocytes, including inflammatory M1 macrophages, mainly shift from oxidative phosphorylation to glycolysis, whereas regulatory T cells and M2 macrophages preferentially use the tricarboxylic acid (TCA) cycle and have reduced glycolysis. Recent studies have revealed the "non-metabolic" signaling functions of intermediates of the mitochondrial pathway and glycolysis. The roles of citrate, succinate and itaconate in immune response, including post-translational modifications of proteins and macrophages activation, have been highlighted. As an end product of glycolysis, lactate has received considerable interest from researchers. In this review, we specifically focused on studies exploring the integration of lactate into immune cell biology and associated pathologies. Lactate can act as a double-edged sword. On one hand, activated immune cells prefer to use lactate to support their function. On the other hand, accumulated lactate in the tissue microenvironment acts as a signaling molecule that restricts immune cell function. Recently, a novel epigenetic change mediated by histone lysine lactylation has been proposed. The burgeoning researches support the idea that histone lactylation participates in diverse cellular events. This review describes glycolytic metabolism, including the immunoregulation of metabolites of the TCA cycle and lactate. These latest findings strengthen our understanding on tumor and chronic inflammatory diseases and offer potential therapeutic options.
Collapse
Affiliation(s)
- Lei Ye
- Department of Gastroenterology and Hepatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Shanghai 200001, China
| | - Yi Jiang
- Department of Gastroenterology and Hepatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Shanghai 200001, China
| | - Mingming Zhang
- Department of Gastroenterology and Hepatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Shanghai 200001, China; Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
14
|
Immune-Related Gene Signatures to Predict the Effectiveness of Chemoimmunotherapy in Triple-Negative Breast Cancer Using Exploratory Subgroup Discovery. Cancers (Basel) 2022; 14:cancers14235806. [PMID: 36497286 PMCID: PMC9735620 DOI: 10.3390/cancers14235806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 11/26/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with limited therapeutic options. Although immunotherapy has shown potential in TNBC patients, clinical studies have only demonstrated a modest response. Therefore, the exploration of immunotherapy in combination with chemotherapy is warranted. In this project we identified immune-related gene signatures for TNBC patients that may explain differences in patients' outcomes after anti-PD-L1+chemotherapy treatment. First, we ran the exploratory subgroup discovery algorithm on the TNBC dataset comprised of 422 patients across 24 studies. Secondly, we narrowed down the search to twelve homogenous subgroups based on tumor mutational burden (TMB, low or high), relapse status (disease-free or recurred), tumor cellularity (high, low and moderate), menopausal status (pre- or post) and tumor stage (I, II and III). For each subgroup we identified a union of the top 10% of genotypic patterns. Furthermore, we employed a multinomial regression model to predict significant genotypic patterns that would be linked to partial remission after anti-PD-L1+chemotherapy treatment. Finally, we uncovered distinct immune cell populations (T-cells, B-cells, Myeloid, NK-cells) for TNBC patients with various treatment outcomes. CD4-Tn-LEF1 and CD4-CXCL13 T-cells were linked to partial remission on anti-PD-L1+chemotherapy treatment. Our informatics pipeline may help to select better responders to chemoimmunotherapy, as well as pinpoint the underlying mechanisms of drug resistance in TNBC patients at single-cell resolution.
Collapse
|
15
|
Martín-Otal C, Navarro F, Casares N, Lasarte-Cía A, Sánchez-Moreno I, Hervás-Stubbs S, Lozano T, Lasarte JJ. Impact of tumor microenvironment on adoptive T cell transfer activity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 370:1-31. [PMID: 35798502 DOI: 10.1016/bs.ircmb.2022.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Recent advances in immunotherapy have revolutionized the treatment of cancer. The use of adoptive cell therapies (ACT) such as those based on tumor infiltrating lymphocytes (TILs) or genetically modified cells (transgenic TCR lymphocytes or CAR-T cells), has shown impressive results in the treatment of several types of cancers. However, cancer cells can exploit mechanisms to escape from immunosurveillance resulting in many patients not responding to these therapies or respond only transiently. The failure of immunotherapy to achieve long-term tumor control is multifactorial. On the one hand, only a limited percentage of the transferred lymphocytes is capable of circulating through the bloodstream, interacting and crossing the tumor endothelium to infiltrate the tumor. Metabolic competition, excessive glucose consumption, the high level of lactic acid secretion and the extracellular pH acidification, the shortage of essential amino acids, the hypoxic conditions or the accumulation of fatty acids in the tumor microenvironment (TME), greatly hinder the anti-tumor activity of the immune cells in ACT therapy strategies. Therefore, there is a new trend in immunotherapy research that seeks to unravel the fundamental biology that underpins the response to therapy and identifies new approaches to better amplify the efficacy of immunotherapies. In this review we address important aspects that may significantly affect the efficacy of ACT, indicating also the therapeutic alternatives that are currently being implemented to overcome these drawbacks.
Collapse
Affiliation(s)
- Celia Martín-Otal
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Flor Navarro
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Noelia Casares
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Aritz Lasarte-Cía
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Inés Sánchez-Moreno
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Sandra Hervás-Stubbs
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Teresa Lozano
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.
| | - Juan José Lasarte
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
| |
Collapse
|
16
|
Augoff K, Hryniewicz-Jankowska A, Tabola R, Stach K. MMP9: A Tough Target for Targeted Therapy for Cancer. Cancers (Basel) 2022; 14:cancers14071847. [PMID: 35406619 PMCID: PMC8998077 DOI: 10.3390/cancers14071847] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Having the capability to proteolyze diverse structural and signaling proteins, matrix metalloproteinase 9 (MMP9), one of the best-studied secretory endopeptidases, has been identified as a crucial mediator of processes closely associated with tumorigenesis, such as the extracellular matrix reorganization, epithelial to mesenchymal transition, cell migration, new blood vessel formation, and immune response. In this review, we present the current state of knowledge on MMP9 and its role in cancer growth in the context of cell adhesion/migration, cancer-related inflammation, and tumor microenvironment formation. We also summarize recent achievements in the development of selective MMP9 inhibitors and the limitations of using them as anticancer drugs.
Collapse
Affiliation(s)
- Katarzyna Augoff
- Department of Surgical Education, Wroclaw Medical University, 50-367 Wroclaw, Poland
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
- Correspondence:
| | | | - Renata Tabola
- Department of Thoracic Surgery, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Kamilla Stach
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| |
Collapse
|
17
|
Zeng Z, Liu Y, Wen Q, Li Y, Yu J, Xu Q, Wan W, He Y, Ma C, Huang Y, Yang H, Jiang O, Li F. Experimental study on preparation and anti-tumor efficiency of nanoparticles targeting M2 macrophages. Drug Deliv 2021; 28:943-956. [PMID: 33988472 PMCID: PMC8128207 DOI: 10.1080/10717544.2021.1921076] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/08/2023] Open
Abstract
This study aimed to develop an effective therapy against M2 macrophages and to investigate the effects of imidazole and mannose modified carboxymethyl chitosan-nanoparticles (MIC-NPs) on tumor growth and antitumor immune responses. MIC-NPs were constructed and analyzed through 1H NMR, nano-laser particle size analyzer, and transmission electron microscopy. The nanoparticles were mainly distributed in 75-85 nm, and zeta potential was 1.5 mV. Cytotoxicity studies in vitro and in vivo indicated that MIC-NPs were safe. The targeting effect of MIC-NPs on M2 macrophages was observed through fluorescence microscope and microplate system. The results demonstrated the uptake of a large amount of FITC-loaded MIC-NPs by M2. Cell growth inhibition experiments showed that MIC-NPs significantly inhibited M2 through cell apoptosis. The evaluation of anti-tumor activity in vivo showed that MIC-NPs could accumulate in the tumor site to exert an anti-tumor effect. Flow cytometry showed that the proportion of M2 macrophages at the tumor site in the experimental group was significantly lower than that in the control group, while the Treg cells and cytotoxic T cells (CTL) were found to be increased. PCR detection showed that the cDNA of FIZZ, MR, TGF-β, and arginase, closely related to M2 macrophages, in the experimental group, was significantly lower than that in the control group, but there was no significant difference in the cDNA of Treg cell characteristic Foxp3 between the two groups. These results suggest that MIC-NPs are expected to provide a new and effective treatment for tumor.
Collapse
Affiliation(s)
- Zheng Zeng
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yu Liu
- Department of Surgical Oncology, The Second People's Hospital of Neijiang, Neijiang, China
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yixian Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jing Yu
- Department of Surgical Oncology, The Second People's Hospital of Neijiang, Neijiang, China
| | - Qiang Xu
- Department of Surgical Oncology, The Second People's Hospital of Neijiang, Neijiang, China
| | - Wenwu Wan
- Clinical Medical College of Southwest Medical University, Luzhou, China
| | - Yu He
- Department of Oncology, The Fourth People's Hospital of Neijiang, Neijiang, China
| | - Chen Ma
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, China
| | - Yan Huang
- Department of Surgical Oncology, The Second People's Hospital of Neijiang, Neijiang, China
| | - Helin Yang
- Department of Surgical Oncology, The Second People's Hospital of Neijiang, Neijiang, China
| | - Ou Jiang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Surgical Oncology, The Second People's Hospital of Neijiang, Neijiang, China
| | - Fuyu Li
- Department of Biliary Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Probiotics in Counteracting the Role of Neutrophils in Cancer Metastasis. Vaccines (Basel) 2021; 9:vaccines9111306. [PMID: 34835236 PMCID: PMC8621509 DOI: 10.3390/vaccines9111306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 12/20/2022] Open
Abstract
Neutrophils are known for their role geared towards pathogen clearance by different mechanisms that they initiate, primarily by the release of neutrophil extracellular traps (NETs). However, their immune-surveillance capacity accompanied with plasticity in existing as interchangeable subsets, discovered recently, has revealed their property to contribute to complex cancer pathologies including tumor initiation, growth, angiogenesis and metastasis. Although there is a growing body of evidence suggesting a critical balance between the protumoral and antitumoral neutrophil phenotypes, an in-depth signaling pathway analysis would aid in determination of anticipatory, diagnostic and therapeutic targets. This review presents a comprehensive overview of the potential pathways involved in neutrophil-triggered cancer metastasis and introduces the influence of the microbial load and avenues for probiotic intervention.
Collapse
|
19
|
Wang J, Wang X, Guo Y, Ye L, Li D, Hu A, Cai S, Yuan B, Jin S, Zhou Y, Li Q, Zheng L, Tong Q. Therapeutic targeting of SPIB/SPI1-facilitated interplay of cancer cells and neutrophils inhibits aerobic glycolysis and cancer progression. Clin Transl Med 2021; 11:e588. [PMID: 34841706 PMCID: PMC8567044 DOI: 10.1002/ctm2.588] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/24/2021] [Accepted: 09/14/2021] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND As a metabolic reprogramming feature, cancer cells derive most of their energy from aerobic glycolysis, while its regulatory mechanisms and therapeutic strategies continue to be illusive. METHODS Integrative analysis of publically available expression profile datasets was used to identify critical transcriptional regulators and their target glycolytic enzymes. The functions and acting mechanisms of transcriptional regulators in cancer cells were investigated by using in vitro and in vivo assays. The Kaplan-Meier curve and log-rank assay were used to conduct the survival study. RESULTS Salmonella pathogenicity island 1 (SPI1/PU.1), a haematopoietic transcription factor, was identified to facilitate glycolytic process, tumourigenesis, invasiveness, as well as metastasis of colon cancer cells, which was interplayed by tumour-associated neutrophils. Mechanistically, neutrophils delivered SPI1 mRNA via extracellular vesicles, resulting in enhanced SPI1 expression of cancer cells. Through physical interaction with SPI1-related protein (SPIB), SPI1 drove expression of glycolytic genes within cancer cells, which in turn induced polarization of neutrophils via glycolytic metabolite lactate. Depletion of neutrophils or SPIB-SPI1 interaction in cancer cells significantly inhibited glycolytic process, tumourigenesis and aggressiveness. Upregulation of SPI1 or SPIB was found to be associated with poor prognosis in patients suffering from colon cancer. CONCLUSIONS Therapeutic targeting of SPIB/SPI1-facilitated interplay of cancerous cells and neutrophils suppresses aerobic glycolysis and progression of cancer.
Collapse
Affiliation(s)
- Jianqun Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Xiaojing Wang
- Department of Geriatrics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Yanhua Guo
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Lin Ye
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Dan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Anpei Hu
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Shuang Cai
- Department of Pathology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Boling Yuan
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Shikai Jin
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Yi Zhou
- Department of Pathology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Qilan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| |
Collapse
|
20
|
Tong N, He Z, Ma Y, Wang Z, Huang Z, Cao H, Xu L, Zou Y, Wang W, Yi C, Yin Z, Wang Q. Tumor Associated Macrophages, as the Dominant Immune Cells, Are an Indispensable Target for Immunologically Cold Tumor-Glioma Therapy? Front Cell Dev Biol 2021; 9:706286. [PMID: 34368156 PMCID: PMC8337013 DOI: 10.3389/fcell.2021.706286] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor microenvironment (TME) is the cornerstone of the occurrence, development, invasion and diffusion of the malignant central nerve system (CNS) tumor, glioma. As the largest number of inflammatory cells in glioma TME, tumor associated macrophages (TAMs) and their secreted factors are indispensable to the progression of glioma, which is a well-known immunologically “cold” tumor, including the growth of tumor cells, invasion, migration, angiogenesis, cancer immunosuppression and metabolism. TAMs intimately interface with the treatment failure and poor prognosis of glioma patients, and their density increases with increasing glioma grade. Recently, great progress has been made in TAM-targeting for anti-tumor therapy. According to TAMs’ function in tumorigenesis and progression, the major anti-tumor treatment strategies targeting TAMs are to hinder macrophage recruitment in TME, reduce TAMs viability or remodel TAMs phenotype from M2 to M1. Different approaches offer unique and effective anti-tumor effect by regulating the phagocytosis, polarization and pro-tumor behaviors of macrophages in the therapy of glioma. The present review summarizes the significant characteristics and related mechanisms of TAMs and addresses the related research progress on targeting TAMs in glioma.
Collapse
Affiliation(s)
- Ni Tong
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhenqiang He
- State Key Laboratory of Oncology in South China, Department of Neurosurgery/Neuro-Oncology, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yujie Ma
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zheng Wang
- Breast Surgery Department, Nanyang Central Hospital, Nanyang, China
| | - Ziming Huang
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Haihong Cao
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lanyang Xu
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuheng Zou
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wanyu Wang
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chenpeng Yi
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhixin Yin
- School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Qirui Wang
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
21
|
Intra-nasal administration of sperm head turns neutrophil into reparative mode after PGE1- and/or Ang II receptor-mediated phagocytosis followed by expression of sperm head's coding RNA. Int Immunopharmacol 2021; 98:107696. [PMID: 34147914 DOI: 10.1016/j.intimp.2021.107696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/25/2021] [Accepted: 04/18/2021] [Indexed: 01/01/2023]
Abstract
Having played homeostatic role, the immune system maintains the integrity of the body. Such a characteristic makes immune system as an attractive candidate for resolution of inflammatory disease followed by tissue repair. As first responder cells, neutrophils direct immune response playing key role in tissue remodeling. Previous studies revealed that sperm attracts neutrophils and promotes uterine remodeling suitable for fetus growth. Accordingly, sperm and more efficiently sperm head had remodeling effects on damaged brain in Alzheimer's disease (AD) model. To further reveal the mechanism, two kinds of in vivo study, including kinetic study and inhibition of neutrophil phagocytosis on AD model, as well as in vitro study using co-culture of neutrophil and sperm head were performed. Kinetic study revealed that sperm head recruited neutrophil to nasal mucosa similar to that of uterus and sperm head-phagocytizing neutrophils acquired new activation status comparing to control. In vitro study also demonstrated that sperm head-phagocytizing neutrophils acquire new activation status and express coding RNAs of sperm head. Accordingly, inhibition of neutrophil phagocytic activity abrogated therapeutic effects of sperm head. Neutrophils activation status is important in the fate of inflammatory process. Modulation but not suppression of neutrophils helps remodeling and repair of damaged tissue. Sperm head is an intelligent cell and not just a simple particle to remove by phagocytosis but instead can program neutrophils and consequently immune response into reparative mode after phagocytosis.
Collapse
|
22
|
Kalinina A, Golubeva I, Kudryavtsev I, Khromova N, Antoshina E, Trukhanova L, Gorkova T, Kazansky D, Khromykh L. Cyclophilin A is a factor of antitumor defense in the early stages of tumor development. Int Immunopharmacol 2021; 94:107470. [PMID: 33640856 DOI: 10.1016/j.intimp.2021.107470] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/14/2021] [Accepted: 02/02/2021] [Indexed: 01/12/2023]
Abstract
Cyclophilin A (CypA) is a pro-inflammatory factor with multiple immunomodulating effects. Here, we investigated the effects of recombinant human CypA (rhCypA) as a factor of antitumor host defense. Our results demonstrated that rhCypA dramatically inhibited the growth of murine transplantable tumors (mammary adenocarcinoma Ca755, melanoma B16, Lewis lung carcinoma (LLC), and cervical cancer CC-5). In the B16 model, rhCypA effects were observed only when tumor cells were transplanted at the significantly reduced injection dose, indicating that antitumor properties of rhCypA are more effective at the initial stages of cancer development. Antitumor effect of rhCypA in the CC-5 model was comparable to the action of 5-fluorouracil (5FU), and rhCypA administration prevented 5FU - induced leukopenia in the blood of tumor-bearing mice. In the LLC model, rhCypA injection before but not after tumor resection significantly suppressed the formation of post-surgical metastases. RhCypA exhibited no direct cytotoxic effects in vitro on human leukemia cells (K-562, HL-60, KG-1), indicating that rhCypA antitumor action could be mediated by its immunomodulating activity. In the B16 model, rhCypA had no impact on tumor angiogenesis and gene expression of several MMPs, endogenous CypA, and CD147, which play a crucial role in cancer progression. However, in this model, rhCypA stimulated gene expression of MMPs 8, 9, and 12 that could contribute to malignancy growth inhibition. Here, our findings pointed out CypA as one of the factors of antitumor host defense that can effectively control the initial stages of tumor and metastases formation by regulating the action of MMPs and changing the tumor microenvironment.
Collapse
Affiliation(s)
- Anastasiia Kalinina
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Irina Golubeva
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Igor Kudryavtsev
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Natalia Khromova
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Elena Antoshina
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Lubov Trukhanova
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Tatyana Gorkova
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Dmitry Kazansky
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Ludmila Khromykh
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation.
| |
Collapse
|
23
|
In-silico modeling and analysis of the therapeutic potential of miRNA-7 on EGFR associated signaling network involved in breast cancer. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Chen Y, Wang Q, Li Z, Liu Z, Zhao Y, Zhang J, Liu M, Wang Z, Li D, Han J. Naproxen platinum(iv) hybrids inhibiting cycloxygenases and matrix metalloproteinases and causing DNA damage: synthesis and biological evaluation as antitumor agents in vitro and in vivo. Dalton Trans 2020; 49:5192-5204. [PMID: 32236281 DOI: 10.1039/d0dt00424c] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cycloxygenases (COXs) and matrix metalloproteinases (MMPs) in the tumor microenvironment (TME) are tightly related to the progression of cancers. Here, naproxen as a potent inhibitor of both COX and MMP was combined with platinum(iv) to construct hybrids as antitumor agents. Compound 2 with comparable or even superior activities to that of cisplatin, oxaliplatin, and carboplatin, great potential for reversing drug resistance, and superior tumor targeting properties was screened out as a lead compound. Moreover, compound 2 possessed potent tumor growth inhibition capability in vivo, which was comparable to that of oxaliplatin, and displayed rather lower side effects than the platinum(ii) reference drugs. The naproxen platinum(iv) complex could easily undergo reduction and liberate the platinum(ii) complex and naproxen as well as exert a multifunctional antitumor mechanism: (i) the liberated platinum(ii) fragment would cause serious DNA injury; (ii) naproxen would inhibit COX-2 and decrease tumor-associated inflammation; and (iii) the naproxen platinum(iv) complex exhibited remarkable MMP-9 inhibition in tumor tissues. These antitumor functions can help reduce the growth and metastasis of malignancy.
Collapse
Affiliation(s)
- Yan Chen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China.
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China.
| | - Zuojie Li
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China.
| | - Zhifang Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China.
| | - Yanna Zhao
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China.
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining 272067, P.R. China
| | - Min Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China.
| | - Zhengping Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China.
| | - Dacheng Li
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China. and Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, Liaocheng University, Liaocheng 252059, PR China
| | - Jun Han
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China.
| |
Collapse
|
25
|
Gordon-Weeks A, Yuzhalin AE. Cancer Extracellular Matrix Proteins Regulate Tumour Immunity. Cancers (Basel) 2020; 12:E3331. [PMID: 33187209 PMCID: PMC7696558 DOI: 10.3390/cancers12113331] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
The extracellular matrix (ECM) plays an increasingly recognised role in the development and progression of cancer. Whilst significant progress has been made in targeting aspects of the tumour microenvironment such as tumour immunity and angiogenesis, there are no therapies that address the cancer ECM. Importantly, immune function relies heavily on the structure, physics and composition of the ECM, indicating that cancer ECM and immunity are mechanistically inseparable. In this review we highlight mechanisms by which the ECM shapes tumour immunity, identifying potential therapeutic targets within the ECM. These data indicate that to fully realise the potential of cancer immunotherapy, the cancer ECM requires simultaneous consideration.
Collapse
Affiliation(s)
- Alex Gordon-Weeks
- Nuffield Department of Surgical Sciences, University of Oxford, Room 6607, Level 6 John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Arseniy E. Yuzhalin
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| |
Collapse
|
26
|
Azevedo Martins JM, Rabelo-Santos SH, do Amaral Westin MC, Zeferino LC. Tumoral and stromal expression of MMP-2, MMP-9, MMP-14, TIMP-1, TIMP-2, and VEGF-A in cervical cancer patient survival: a competing risk analysis. BMC Cancer 2020; 20:660. [PMID: 32669083 PMCID: PMC7364527 DOI: 10.1186/s12885-020-07150-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/07/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Expression of matrix metalloproteases 2, 9 and 14 (MMP-2, MMP-9, MMP-14), tissue inhibitors of metalloprotease 1 and 2 (TIMP-1, TIMP-2) and vascular endothelial growth factor A (VEGF-A) is involved in tumor invasion and metastasis via extracellular matrix degradation and angiogenesis. This study aimed to assess whether the expression of MMP-2, MMP-9, MMP-14, TIMP-1, and TIMP-2 in tumors and in the adjacent stroma is associated with cervical cancer prognosis. METHODS This study analyzed a retrospective cohort of 64 patients. Protein expression was previously obtained by immunohistochemistry from biopsies containing both tumor and stroma. The expression and percentage of stained cells were categorized as high or low according to the cutoff points by using ROC curves. The follow-up data was collected from diagnosis to the last clinical visit. Clinical status categorized as alive without disease, alive with disease, death due to other causes, and death from the disease. The relative risk of death from the disease was evaluated according to the proteins expression using a cause-specific Cox regression model with a 95% confidence interval (95%CI). For the significant associations (p < 0.05), survival curves of patients with low and high expression were plotted for the competing risk survival curve analyses. RESULTS High expression levels of stromal MMP-2 (RR; 95%CI: 3.91; 1.17-13.02) and stromal TIMP-2 (RR, 95%CI: 8.67; 1.15-65.27) were associated with a greater relative risk of death from the disease and with lower survival (p = 0.03; p = 0.04) than lower expression levels. Low expression levels of stromal MMP-9 (RR, 95%CI: 0.19; 0.05-0.65) and tumoral MMP-9 (HR, 95%CI: 0.19; 0.04-0.90) were protective factors against death from the disease and were associated with poorer survival. CONCLUSIONS High expression levels of MMP-2 and TIMP-2 in the stroma were significantly associated with poor survival in cervical cancer patients. High expression of MMP-9 was associated with a favorable cervical cancer prognosis.
Collapse
Affiliation(s)
- Jordana Maria Azevedo Martins
- Department of Gynecology and Obstetrics, School of Medical Sciences, State University of Campinas, UNICAMP, Tessalia Vieira de Camargo Street, 126, Campinas, Sao Paulo 13083-887 Brazil
| | - Silvia Helena Rabelo-Santos
- School of Pharmacy, Federal University of Goias, 240 Street, Leste Universitario, Goiania, Goias 74605-170 Brazil
| | - Maria Cristina do Amaral Westin
- Laboratory of Cytopathology, Women’s Health Hospital Professor Jose Aristodemo Pinotti – (CAISM), University of Campinas (UNICAMP), Campinas, Sao Paulo 13083-881 Brazil
| | - Luiz Carlos Zeferino
- Department of Gynecology and Obstetrics, School of Medical Sciences, State University of Campinas, UNICAMP, Tessalia Vieira de Camargo Street, 126, Campinas, Sao Paulo 13083-887 Brazil
| |
Collapse
|
27
|
Molecular evaluation of chronic restrain stress in mice model of non metastatic fibrosarcoma. J Mol Histol 2020; 51:367-374. [PMID: 32556790 DOI: 10.1007/s10735-020-09886-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/12/2020] [Indexed: 01/09/2023]
Abstract
Chronic stress is regarded as a significant factor in the etiology of the many diseases. Numerous methods have been developed through which the effect of chronic stress is examined. The aim of this study is to demonstrate the new experimental model for analysis of immuno-suppression induced by chronic restraint stress, through challenge with conditionally tumorigenic cell line BHK-21/C13. 20 male NMRI mice were randomly divided into 2 groups-control and experimental. Each mouse was subcutaneously inoculated with BHK-21/C13 cells. Stress in the experimental group was induced for 20 days. After the experiment, tumor masses were removed, and analyzed using histology and immunohistochemistry techniques. We found a statistically significant difference (p = 0.034) in tumor expression and tumor volumes (p = 0.0061) between groups, as well as in immunopositivity on Ki67, cytochrome C and matrix metalloproteinase 9. Absence of immune infiltrate was noticed in experimental, and the presence of inflammatory infiltrate at tumor invasion front in control group.
Collapse
|
28
|
Panda SP, Panigrahy UP, Prasanth D, Gorla US, Guntupalli C, Panda DP, Jena BR. A trimethoxy flavonoid isolated from stem extract of Tabebuia chrysantha suppresses angiogenesis in angiosarcoma. J Pharm Pharmacol 2020; 72:990-999. [PMID: 32311118 DOI: 10.1111/jphp.13272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/21/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVES This research aimed to evaluate the antiangiogenic activity of isolated flavonoid 4a,5,8,8a-tetrahydro-5-hydroxy-3,7,8-trimethoxy-2-(3,4-dimethoxyphenyl) chromen-4-one (TMF) from Tabebuia chrysantha. STAT3-MMP9 signalling is a signal transduction mechanism that promotes angiogenesis in various cancers. METHODS The tumour xenografting chicken embryo chorioallantoic membrane (CAM) model-based ex vivo assay was used to evaluate the activity of TMF. The Western blot, densitometric analysis and quantitative real-time polymerase chain reaction (qRT-PCR) were performed to evaluate the activity of the MMP9. Zebrafish embryos were used to evaluate embryotoxicity, and in vitro free radical scavenging activity of flavonoid was also elucidated. KEY FINDINGS This research assessed the high level of STAT3, p-ERK, VEGF-R and MMP9 in the tissue extract of the control group, and also, the suppression of angiogenesis in the treatment groups was due to scavenged ROS and RNS, dephosphorylation of STAT3 and ERK, and suppression of MMP9 gene expression. CONCLUSION The isolated flavonoid named TMF from T. chrysantha functions as specific regulators of target proteins of angiosarcoma. The STAT3-MMP9 signalling may be used as an effective prognostic marker of angiosarcoma.
Collapse
Affiliation(s)
- Siva Prasad Panda
- Pharmacology research division, College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India
| | | | - Dsnbk Prasanth
- Pharmacology research division, College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India
| | - Uma Sankar Gorla
- Pharmacology research division, College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India
| | - Chakravarthi Guntupalli
- Pharmacology research division, College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India
| | | | - Bikash Ranjan Jena
- Southern Institute of Medical Sciences (SIMS college of Pharmacy), Guntur, India
| |
Collapse
|
29
|
Shamloo B, Kumar N, Owen RH, Reemmer J, Ost J, Perkins RS, Shen HY. Dysregulation of adenosine kinase isoforms in breast cancer. Oncotarget 2019; 10:7238-7250. [PMID: 31921385 PMCID: PMC6944449 DOI: 10.18632/oncotarget.27364] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/07/2019] [Indexed: 12/25/2022] Open
Abstract
Dysregulated adenosine signaling pathway has been evidenced in the pathogenesis of breast cancer. However, the role of adenosine kinase (ADK) in tumorigenesis remains unclear while it crucially regulates the removal and availability of adenosine. ADK has two isoforms that localize to discrete subcellular spaces: i.e., nuclear, long-isoform (ADK-L) and cytosolic, short-isoform (ADK-S). We hypothesized that these two ADK isoforms would be differentially expressed in breast cancer and may contribute to divergent cellular actions in cancer. In this study, we examined the expression profiles of ADK isoforms in breast cancer tissues from 46 patient and followed up with an in vitro investigation by knocking down the expression of ADK-L or ADK-S using CRISPR gene editing to evaluate the role of ADK isoform in cancer progression and metastasis of cultured triple-negative breast cancer cell line MDA-MB-231. We demonstrated that (i) ADK-L expression level was significantly increased in breast cancer tissues versus paired normal tissues adjacent to tumor, whereas the ADK-S expression levels were not significantly different between cancerous and normal tissues; (ii) CRISPR/Cas9-mediated downregulation of ADK isoforms, led to suppressed cellular proliferation, division, and migration of cultured breast cancer cells; (iii) ADK-L knockdown significantly upregulated gene expression of matrix metalloproteinase (ADAM23, 9.93-fold; MMP9, 24.58-fold) and downregulated expression of cyclin D2 (CCND2, -30.76-fold), adhesive glycoprotein THBS1 (-8.28-fold), and cystatin E/M (CST6, -16.32-fold). Our findings suggest a potential role of ADK-L in mitogenesis, tumorigenesis, and tumor-associated tissue remodeling and invasion; and the manipulation of ADK-L holds promise as a therapeutic strategy for aggressive breast cancer.
Collapse
Affiliation(s)
- Bahar Shamloo
- Department of Translational Neuroscience, Legacy Research Institute, Legacy Health, Portland, OR 97232, USA
| | - Nandita Kumar
- Department of Translational Neuroscience, Legacy Research Institute, Legacy Health, Portland, OR 97232, USA
| | - Randall H Owen
- Department of Translational Neuroscience, Legacy Research Institute, Legacy Health, Portland, OR 97232, USA
| | - Jesica Reemmer
- Department of Translational Neuroscience, Legacy Research Institute, Legacy Health, Portland, OR 97232, USA
| | - John Ost
- Legacy Tumor Bank, Legacy Research Institute, Legacy Health, Portland, OR 97232, USA
| | - R Serene Perkins
- Legacy Tumor Bank, Legacy Research Institute, Legacy Health, Portland, OR 97232, USA.,Mid-Columbia Medical Center, The Dalles, OR 97058, USA
| | - Hai-Ying Shen
- Department of Translational Neuroscience, Legacy Research Institute, Legacy Health, Portland, OR 97232, USA
| |
Collapse
|
30
|
Lindahl G, Rzepecka A, Dabrosin C. Increased Extracellular Osteopontin Levels in Normal Human Breast Tissue at High Risk of Developing Cancer and Its Association With Inflammatory Biomarkers in situ. Front Oncol 2019; 9:746. [PMID: 31475105 PMCID: PMC6707004 DOI: 10.3389/fonc.2019.00746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/25/2019] [Indexed: 01/13/2023] Open
Abstract
Mammographic breast density is a strong independent risk factor for breast cancer (BC), but the molecular mechanisms behind this risk is yet undetermined and prevention strategies for these women are lacking. The anti-estrogen tamoxifen may reduce the risk of BC but this treatment is associated with severe side effects. Thus, other means for BC prevention, such as diet interventions, need to be developed. Osteopontin (OPN) is a major mediator of inflammation which is key in carcinogenesis. OPN may be cleaved by proteases in the tissue and cleaved OPN may in turn induce an inflammatory cascade in the extracellular microenvironment. We aimed to determine if extracellular OPN was altered in BC and in normal breast tissue with different densities and if tamoxifen or a diet of flaxseed could modify OPN levels. The study comprised 103 women; 13 diagnosed with BC, 42 healthy post-menopausal women with different breast densities at their mammography screen, and 34 post-menopausal women who added 25 g of ground flaxseed/day or were treated with tamoxifen 20 mg/day and were investigated before and after 6 weeks of exposure. Additionally, 10 premenopausal women who added flaxseed for one menstrual cycle and four who were investigated in two unexposed consecutive luteal phases of the menstrual cycle. Microdialysis was used to sample extracellular proteins in vivo in breast tissue and proteins were quantified using a multiplex proximity extension assay. We found that, similar to BC, extracellular in vivo OPN levels were significantly increased in dense breast tissue. Additionally, significant correlations were found between OPN and chemokine (C-X-C motif) ligand (CXCL)-1, −8, −9, −10, and −11, interleukin-6, vascular endothelial growth factor, matrix metalloproteinase (MMP)-1, −2, −3, 7, and −12 and urokinase-type plasminogen activator whereas no correlations were found with MMP-9, chemokine (C-C motif) ligand (CCL)-2, and −5. Estradiol did not affect OPN levels in breast tissue. None of the interventions altered OPN levels. The pro-tumorigenic protein OPN may indeed be a molecular target for BC prevention in women with increased breast density but other means than tamoxifen or flaxseed i.e., more potent anti-inflammatory approaches, need to be evaluated for this purpose.
Collapse
Affiliation(s)
- Gabriel Lindahl
- Department of Oncology, Linköping University, Linköping, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Anna Rzepecka
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Department of Radiology, Linköping University, Linköping, Sweden.,Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Charlotta Dabrosin
- Department of Oncology, Linköping University, Linköping, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
31
|
Innate immune cell infiltration in melanoma metastases affects survival and is associated with BRAFV600E mutation status. Melanoma Res 2019; 29:30-37. [PMID: 30299387 DOI: 10.1097/cmr.0000000000000515] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Little is known about the infiltrative pattern of innate immune cells in primary melanoma compared with their paired metastases and in BRAF-mutated tumors. Therefore, our aim was to characterize the inflammatory microenvironment in primary ulcerated and nonulcerated melanomas and paired metastases, to investigate the relation between inflammation and BRAF mutation in primary melanoma and paired metastases, and to evaluate the effect of the analyzed biomarkers on melanoma-specific survival. A total of 385 primary tumors and 96 paired metastases were stained with immunohistochemistry for BRAF, CD163+ macrophages, CD123+ plasmacytoid dendritic cells, CD66b+ neutrophils, and E-cadherin and estimated using objective computer-assisted image analysis. BRAF was semiquantitatively scored as either present or absent. In metastases of nonulcerated melanomas, we observed higher neutrophil (P=0.02) and macrophage (P=0.01) numbers. In the metastases of ulcerated melanomas, we found a higher number of macrophages (P<0.0001). Increase in the neutrophil numbers in the metastases was associated with poor patient survival after first relapse (hazard ratio=1.19, 95% confidence interval: 1.03-1.38, P=0.02). BRAF-positive primary tumors (P=0.02) and metastases (P=0.01) exhibited increased plasmacytoid dendritic cell numbers compared with BRAF-negative tumors. Lastly, primary melanomas in men had higher neutrophil numbers than women (P≤0.0001), and men had worse melanoma-specific survival (hazard ratio=1.52, 95% confidence interval: 1.04-2.21, P=0.03). Our data show that melanoma metastases are densely infiltrated with neutrophils, which affects survival. Our results also highlight the importance of recognizing the presence of inflammatory cells in the metastases as a prognostic marker, and that they may potentially be used to improve the precision of immunotherapy and BRAF targeted therapy.
Collapse
|
32
|
Ahmad N, Colak B, Gibbs MJ, Zhang DW, Gautrot JE, Watkinson M, Becer CR, Krause S. Peptide Cross-Linked Poly(2-oxazoline) as a Sensor Material for the Detection of Proteases with a Quartz Crystal Microbalance. Biomacromolecules 2019; 20:2506-2514. [PMID: 31244015 DOI: 10.1021/acs.biomac.9b00245] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Inflammatory conditions are frequently accompanied by increased levels of active proteases, and there is rising interest in methods for their detection to monitor inflammation in a point of care setting. In this work, new sensor materials for disposable single-step protease biosensors based on poly(2-oxazoline) hydrogels cross-linked with a protease-specific cleavable peptide are described. The performance of the sensor material was assessed targeting the detection of matrix metalloproteinase-9 (MMP-9), a protease that has been shown to be an indicator of inflammation in multiple sclerosis and other inflammatory conditions. Films of the hydrogel were formed on gold-coated quartz crystals using thiol-ene click chemistry, and the cross-link density was optimized. The degradation rate of the hydrogel was monitored using a quartz crystal microbalance (QCM) and showed a strong dependence on the MMP-9 concentration. A concentration range of 0-160 nM of MMP-9 was investigated, and a lower limit of detection of 10 nM MMP-9 was determined.
Collapse
Affiliation(s)
- Norlaily Ahmad
- School of Engineering and Materials Science , Queen Mary University of London , London , E1 4NS , United Kingdom.,Centre of Foundation Studies , Universiti Teknologi MARA , Cawangan Selangor, Kampus Dengkil , 43800 Dengkil , Selangor , Malaysia
| | - Burcu Colak
- School of Engineering and Materials Science , Queen Mary University of London , London , E1 4NS , United Kingdom
| | - Martin John Gibbs
- School of Engineering and Materials Science , Queen Mary University of London , London , E1 4NS , United Kingdom
| | - De-Wen Zhang
- Institute of Medical Engineering, School of Basic Medical Sciences , Xi'an Jiaotong University Health Science Center , Xi'an , 710061 , China
| | - Julien E Gautrot
- School of Engineering and Materials Science , Queen Mary University of London , London , E1 4NS , United Kingdom
| | - Michael Watkinson
- The Lennard-Jones Laboratories, School of Chemical and Physical Sciences , Keele University , Staffordshire , ST5 5BG , United Kingdom
| | - C Remzi Becer
- Department of Chemistry , University of Warwick , Coventry , CV47AL , United Kingdom
| | - Steffi Krause
- School of Engineering and Materials Science , Queen Mary University of London , London , E1 4NS , United Kingdom
| |
Collapse
|
33
|
Väyrynen O, Åström P, Nyberg P, Alahuhta I, Pirilä E, Vilen ST, Aikio M, Heljasvaara R, Risteli M, Sutinen M, Salo T. Matrix metalloproteinase 9 inhibits the motility of highly aggressive HSC-3 oral squamous cell carcinoma cells. Exp Cell Res 2019; 376:18-26. [PMID: 30710501 DOI: 10.1016/j.yexcr.2019.01.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/23/2019] [Accepted: 01/29/2019] [Indexed: 01/01/2023]
Abstract
Pro-tumorigenic activities of matrix metalloproteinase (MMP) 9 have been linked to many cancers, but recently the tumour-suppressing role of MMP9 has also been elucidated. The multifaceted evidence on this subject prompted us to examine the role of MMP9 in the behaviour of oral tongue squamous cell carcinoma (OTSCC) cells. We used gelatinase-specific inhibitor, CTT2, and short hairpin (sh) RNA gene silencing to study the effects of MMP9 on proliferation, motility and invasion of an aggressive OTSCC cell line, HSC-3. We found that the migration and invasion of HSC-3 cells were increased by CTT2 and shRNA silencing of MMP9. Proliferation, in turn, was decreased by MMP9 inhibition. Furthermore, arresten-overexpressing HSC-3 cells expressed increased levels of MMP9, but exhibited decreased motility compared with controls. Interestingly, these cells restored their migratory capabilities by CTT2 inhibition of MMP9. Hence, although higher MMP9 expression could give rise to an increased tumour growth in vivo due to increased proliferation, in some circumstances, it may participate in yet unidentified molecular mechanisms that reduce the cell movement in OTSCC.
Collapse
Affiliation(s)
- Otto Väyrynen
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Pirjo Åström
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Pia Nyberg
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; Biobank Borealis of Northern Finland, Oulu University Hospital, Finland
| | - Ilkka Alahuhta
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Emma Pirilä
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Suvi-Tuuli Vilen
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Mari Aikio
- Oulu Center for Cell-Matrix Research and Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland
| | - Ritva Heljasvaara
- Oulu Center for Cell-Matrix Research and Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland; Centre for Cancer Biomarkers (CCBIO), University of Bergen, Norway
| | - Maija Risteli
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Meeri Sutinen
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland; HUSLAB, Department of Pathology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
34
|
Lindahl G, Abrahamsson A, Dabrosin C. Dietary flaxseed and tamoxifen affect the inflammatory microenvironment in vivo in normal human breast tissue of postmenopausal women. Eur J Clin Nutr 2019; 73:1250-1259. [DOI: 10.1038/s41430-019-0396-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/18/2018] [Accepted: 12/23/2018] [Indexed: 12/27/2022]
|
35
|
The Relationship Between Phospho-p38, Matrix Metalloproteinase 9, and Major Histocompatibility Complex Class I Chain-Related Molecule A Expression in Pituitary Adenomas Demonstrates a New Mechanism of Pituitary Adenoma Immune Escape. World Neurosurg 2018; 123:e116-e124. [PMID: 30458325 DOI: 10.1016/j.wneu.2018.11.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND The major histocompatibility complex class I chain-related molecule A (MICA) is one of the natural killer group 2D ligands. Soluble major histocompatibility complex class I chain-related molecule A (sMICA) mediates tumor immune escape, but the mechanism is not fully understood. In this study, we examined the expression of phospho-p38, matrix metalloproteinase 9 (MMP-9), and MICA and their relationships among each other in pituitary adenoma tissues to provide a histologic basis for the mechanism of pituitary adenoma immune escape. METHODS We applied immunohistochemistry, real-time quantitative reverse-transcriptase polymerase chain reaction, and Western blot to detect phospho-p38, MMP-9, and MICA expression at the mRNA and protein levels in pituitary adenoma tissues. Enzyme-linked immunosorbent assay was used to examine the expression levels of MMP-9 and sMICA in peripheral blood serum from patients with pituitary adenoma. RESULTS We found that p38, MICA, and MMP-9 mRNA levels were greater in pituitary adenomas than in normal tissues. The phospho-p38, MMP-9, and MICA proteins were overexpressed in pituitary adenomas, and the expression of MMP-9 and MICA were positively correlated with the expression of phospho-p38. In addition, the serum levels of sMICA and MMP-9 proteins in pituitary adenoma patients were significantly greater than those in normal controls. CONCLUSIONS These findings suggest that activation of the p38/mitogen-activated protein kinase pathway may increase MICA expression and induce MMP-9 expression. MMP-9 is involved in the shedding of sMICA from MICA to promote tumor immune escape. Furthermore, p38/mitogen-activated protein kinase could potentially represent a novel target for inhibiting pituitary adenoma immune escape.
Collapse
|
36
|
Elie BT, Fernández-Gallardo J, Curado N, Cornejo MA, Ramos JW, Contel M. Bimetallic titanocene-gold phosphane complexes inhibit invasion, metastasis, and angiogenesis-associated signaling molecules in renal cancer. Eur J Med Chem 2018; 161:310-322. [PMID: 30368130 DOI: 10.1016/j.ejmech.2018.10.034] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/11/2018] [Accepted: 10/14/2018] [Indexed: 01/08/2023]
Abstract
Following promising recent in vitro and in vivo studies of the anticancer efficacies of heterometallic titanocene-gold chemotherapeutic candidates against renal cancer, we report here on the synthesis, characterization, stability studies and biological evaluation of a new titanocene complex containing a gold-triethylphosphane fragment [(η-C5H5)2TiMe(μ-mba)Au(PEt3)] (4) Titanofin. The compound is more stable in physiological fluid than those previously reported, and it is highly cytotoxic against a line of human clear cell renal carcinoma. We describe here preliminary mechanistic data for this compound and previously reported [(η-C5H5)2TiMe(μ-mba)Au(PPh3)] (2) Titanocref which displayed remarkable activity in an in vivo mouse model. Mechanistic studies were carried out in the human clear cell renal carcinoma Caki-1 line for the bimetallic compounds [(η-C5H5)2TiMe(μ-mba)Au(PR3)] (PR3 = PPh32 Titanocref and PEt34 Titanofin), the two monometallic gold derivatives [Au(Hmba)(PR3)] (PR3 = PPh31 cref; PEt33 fin), titanocene dichloride and Auranofin as controls. These studies indicate that bimetallic compounds Titanocref (2) and Titanofin (4) are more cytotoxic than gold monometallic derivatives (1 and 3) and significantly more cytotoxic than titanocene dichloride while being quite selective. Titanocref (2) and Titanofin (4) inhibit migration, invasion, and angiogenic assembly along with molecular markers associated with these processes such as prometastatic IL(s), MMP(s), TNF-α, and proangiogenic VEGF, FGF-basic. The bimetallic compounds also strongly inhibit the mitochondrial protein TrxR often overexpressed in cancer cells evading apoptosis and also inhibit FOXC2, PECAM-1, and HIF-1α whose overexpression is linked to resistance to genotoxic chemotherapy. In summary, bimetallic titanocene-gold phosphane complexes (Titanocref 2 and Titanofin 4) are very promising candidates for further preclinical evaluations for the treatment of renal cancer.
Collapse
Affiliation(s)
- Benelita T Elie
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA; Biology PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA
| | - Jacob Fernández-Gallardo
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Natalia Curado
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Mike A Cornejo
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Joe W Ramos
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, USA
| | - María Contel
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA; Biology PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA; Chemistry PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA; Biochemistry PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA; Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, USA.
| |
Collapse
|
37
|
Matrix Metalloproteinase-9 (MMP-9) as a Cancer Biomarker and MMP-9 Biosensors: Recent Advances. SENSORS 2018; 18:s18103249. [PMID: 30262739 PMCID: PMC6211011 DOI: 10.3390/s18103249] [Citation(s) in RCA: 424] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/23/2018] [Accepted: 09/25/2018] [Indexed: 12/17/2022]
Abstract
As one of the most widely investigated matrix metalloproteinases (MMPs), MMP-9 is a significant protease which plays vital roles in many biological processes. MMP-9 can cleave many extracellular matrix (ECM) proteins to regulate ECM remodeling. It can also cleave many plasma surface proteins to release them from the cell surface. MMP-9 has been widely found to relate to the pathology of cancers, including but not limited to invasion, metastasis and angiogenesis. Some recent research evaluated the value of MMP-9 as biomarkers to various specific cancers. Besides, recent research of MMP-9 biosensors discovered various novel MMP-9 biosensors to detect this enzyme. In this review, some recent advances in exploring MMP-9 as a biomarker in different cancers are summarized, and recent discoveries of novel MMP-9 biosensors are also presented.
Collapse
|
38
|
Galdiero MR, Marone G, Mantovani A. Cancer Inflammation and Cytokines. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028662. [PMID: 28778871 DOI: 10.1101/cshperspect.a028662] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic inflammation is a well-recognized tumor-enabling capability, which allows nascent tumors to escape immunosurveillance. A number of soluble and cellular inflammatory mediators take part in the various phases of cancer initiation and progression, giving rise to a fatal conspiracy, which is difficult to efficiently overcome. Tumor-associated macrophages (TAMs) are pivotal players of the tumor microenvironment and, because of their characteristic plasticity, can acquire a number of distinct phenotypes and contribute in different ways to the various phases of cancerogenesis. Tumor-associated neutrophils (TANs) are also emerging as important components of the tumor microenvironment, given their unexpected heterogeneity and plasticity. TAMs and TANs are both integrated in cancer-related inflammation and an ever better understanding of their functions can be useful to tailor the use of anticancer therapeutic approaches and patient follow-up.
Collapse
Affiliation(s)
- Maria Rosaria Galdiero
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.,Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), National Research Council (CNR), 80131 Naples, Italy
| | - Alberto Mantovani
- Istituto di Ricovero e Cura a Carattere Scientifo (IRCCS), Istituto Clinico Humanitas, Rozzano, Milan, Italy.,Humanitas University, Rozzano, Milan, Italy
| |
Collapse
|
39
|
Akter H, Yoon JH, Yoo YS, Kang MJ. Validation of Neurotensin Receptor 1 as a Therapeutic Target for Gastric Cancer. Mol Cells 2018; 41:591-602. [PMID: 29794962 PMCID: PMC6030244 DOI: 10.14348/molcells.2018.0025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/13/2018] [Accepted: 03/20/2018] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer is the fifth most common type of malignancy worldwide, and the survival rate of patients with advanced-stage gastric cancer is low, even after receiving chemotherapy. Here, we validated neurotensin receptor 1 (NTSR1) as a potential therapeutic target in gastric cancer. We compared NTSR1 expression levels in sixty different gastric cancer-tissue samples and cells, as well as in other cancer cells (lung, breast, pancreatic, and colon), by assessing NTSR1 expression via semi-quantitative real-time reverse transcription polymerase chain reaction, immunocytochemistry and western blot. Following neurotensin (NT) treatment, we analyzed the expression and activity of matrix metalloproteinase-9 (MMP-9) and further determined the effects on cell migration and invasion via wound-healing and transwell assays. Our results revealed that NTSR1 mRNA levels were higher in gastric cancer tissues than non-cancerous tissues. Both of NTSR1 mRNA levels and expression were higher in gastric cancer cell lines relative to levels observed in other cancer-cell lines. Moreover, NT treatment induced MMP-9 expression and activity in all cancer cell lines, which was significantly decreased following treatment with the NTSR1 antagonist SR48692 or small-interfering RNA targeting NTSR1. Furthermore, NT-mediated metastases was confirmed by observing epithelial-mesenchymal transition markers SNAIL and E-cadherin in gastric cancer cells. NT-mediated invasion and migration of gastric cancer cells were reduced by NTSR1 depletion through the Erk signaling. These findings strongly suggested that NTR1 constitutes a potential therapeutic target for the inhibition of gastric cancer invasion and metastasis.
Collapse
Affiliation(s)
- Hafeza Akter
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792,
Korea
- Department of Biological Chemistry, University of Science and Technology, Daejeon 34113,
Korea
| | - Jung Hwan Yoon
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591,
Korea
| | - Young Sook Yoo
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792,
Korea
| | - Min-Jung Kang
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792,
Korea
- Department of Biological Chemistry, University of Science and Technology, Daejeon 34113,
Korea
| |
Collapse
|
40
|
Pirooz HJ, Jafari N, Rastegari M, Fathi‐Roudsari M, Tasharrofi N, Shokri G, Tamadon M, Sazegar H, Kouhkan F. Functional SNP in microRNA‐491‐5p binding site of MMP9 3′‐UTR affects cancer susceptibility. J Cell Biochem 2018; 119:5126-5134. [DOI: 10.1002/jcb.26471] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/31/2017] [Indexed: 12/24/2022]
Affiliation(s)
| | - Niloofar Jafari
- Department of Biology, Basic Sciences FacultyShahrekord Branch, Islamic Azad UniversityShahrekordIran
| | - Mozhdeh Rastegari
- Department of Biology, Basic Sciences FacultyShahrekord Branch, Islamic Azad UniversityShahrekordIran
| | | | - Nooshin Tasharrofi
- Stem Cell Technology Research CenterTehranIran
- Faculty of PharmacyLorestan University of Medical SciencesKhorramabadIran
- Student Research CommiteeLorestan Universsity of Medical SciencesKhorramabadIran
| | | | | | - Hossein Sazegar
- Department of Biology, Basic Sciences FacultyShahrekord Branch, Islamic Azad UniversityShahrekordIran
| | | |
Collapse
|
41
|
Todorović-Raković N, Radulovic M, Vujasinović T, Milovanović J, Nikolić-Vukosavljević D. The time-dependent prognostic value of intratumoral cytokine expression profiles in a natural course of primary breast cancer with a long-term follow-up. Cytokine 2018; 102:12-17. [DOI: 10.1016/j.cyto.2017.11.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 01/05/2023]
|
42
|
Abrahamsson A, Rzepecka A, Dabrosin C. Equal Pro-inflammatory Profiles of CCLs, CXCLs, and Matrix Metalloproteinases in the Extracellular Microenvironment In Vivo in Human Dense Breast Tissue and Breast Cancer. Front Immunol 2018; 8:1994. [PMID: 29387062 PMCID: PMC5776019 DOI: 10.3389/fimmu.2017.01994] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/22/2017] [Indexed: 12/21/2022] Open
Abstract
The inflammatory microenvironment affects breast cancer progression. Proteins that govern the inflammatory response are secreted into the extracellular space, but this compartment still needs to be characterized in human breast tissues in vivo. Dense breast tissue is a major risk factor for breast cancer by yet unknown mechanisms and no non-toxic prevention for these patients exists. Here, we used the minimal invasive technique of microdialysis for sampling of extracellular proteins in live tissues in situ in breast cancers of women before surgery and in healthy women having dense or non-dense breast tissue on mammography. Proteins were profiled using a proximity extension assay. Out of the 32 proteins assessed, 26 exhibited similar profiles in breast cancers and dense breast tissues; CCL-4, -7, -8, -11, -15, -16, -22, -23, and -25, CXCL-5, -8, -9, -16 as well as sIL-6R, IL-18, vascular endothelial growth factor, TGF-α, fibroblast growth factor 19, matrix metalloproteinase (MMP)-1, -2, -3, and urokinase-type plasminogen activator were all increased, whereas CCL-3, CX3CL1, hepatocyte growth factor, and MMP-9 were unaltered in the two tissues. CCL-19 and -24, CXCL-1 and -10, and IL-6 were increased in dense breast tissue only, whereas IL-18BP was increased in breast cancer only. Our results provide novel insights in the inflammatory microenvironment in human breast cancer in situ and define potential novel therapeutic targets. Additionally, we show previously unrecognized similarities of the pro-inflammatory microenvironment in dense breast tissue and breast cancer in vivo suggesting that anti-inflammatory breast cancer prevention trials for women with dense breast tissue may be feasible.
Collapse
Affiliation(s)
- Annelie Abrahamsson
- Department of Oncology, Linköping University, Linköping, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Anna Rzepecka
- Department of Radiology, Linköping University, Linköping, Sweden.,Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Charlotta Dabrosin
- Department of Oncology, Linköping University, Linköping, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
43
|
Galdiero MR, Varricchi G, Loffredo S, Mantovani A, Marone G. Roles of neutrophils in cancer growth and progression. J Leukoc Biol 2017; 103:457-464. [DOI: 10.1002/jlb.3mr0717-292r] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/15/2017] [Accepted: 10/09/2017] [Indexed: 01/01/2023] Open
Affiliation(s)
- Maria Rosaria Galdiero
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II; Naples Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II; Naples Italy
- WAO Center of Excellence, University of Naples Federico II; Naples Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II; Naples Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II; Naples Italy
- WAO Center of Excellence, University of Naples Federico II; Naples Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II; Naples Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II; Naples Italy
- WAO Center of Excellence, University of Naples Federico II; Naples Italy
| | - Alberto Mantovani
- Humanitas Clinical and Research Center; Rozzano Milan Italy
- Department of Biomedical Sciences; Humanitas University; Milan Italy
| | - Gianni Marone
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II; Naples Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II; Naples Italy
- WAO Center of Excellence, University of Naples Federico II; Naples Italy
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS); National Research Council (CNR); Naples Italy
| |
Collapse
|
44
|
El-Shennawy L, Dubrovskyi O, Kastrati I, Danes JM, Zhang Y, Whiteley HE, Creighton CJ, Frasor J. Coactivation of Estrogen Receptor and IKKβ Induces a Dormant Metastatic Phenotype in ER-Positive Breast Cancer. Cancer Res 2017; 78:974-984. [PMID: 29229606 DOI: 10.1158/0008-5472.can-17-1686] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 11/01/2017] [Accepted: 12/05/2017] [Indexed: 02/07/2023]
Abstract
A growing body of evidence suggests that the inflammatory NFκB pathway is associated with the progression of ER+ tumors to more aggressive stages. However, it is unknown whether NFκB is a driver or a consequence of aggressive ER+ disease. To investigate this question, we developed breast cancer cell lines expressing an inducible, constitutively active form of IκB kinase β (CA-IKKβ), a key kinase in the canonical NFκB pathway. We found that CA-IKKβ blocked E2-dependent cell proliferation in vitro and tumor growth in vivo in a reversible manner, suggesting that IKKβ may contribute to tumor dormancy and recurrence of ER+ disease. Moreover, coactivation of ER and IKKβ promoted cell migration and invasion in vitro and drove experimental metastasis in vivo Gene expression profiling revealed a strong association between ER and CA-IKKβ-driven gene expression and clinically relevant invasion and metastasis gene signatures. Mechanistically, the invasive phenotype appeared to be driven by an expansion of a basal/stem-like cell population rather than EMT. Taken together, our findings suggest that coactivation of ER and the canonical NFκB pathway promotes a dormant, metastatic phenotype in ER+ breast cancer and implicates IKKβ as a driver of certain features of aggressive ER+ breast cancer.Significance: The canonical NFκB pathway promotes expansion of stem/basal-like cells and a dormant, metastatic phenotype in ER+ breast cancer cells. Cancer Res; 78(4); 974-84. ©2017 AACR.
Collapse
Affiliation(s)
- Lamiaa El-Shennawy
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois.,Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Oleksii Dubrovskyi
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Irida Kastrati
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Jeanne M Danes
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Herbert E Whiteley
- College of Veterinary Medicine, University of Illinois, Urbana, Illinois
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Jonna Frasor
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
45
|
Abstract
Human cancers exhibit formidable molecular heterogeneity, to a large extent accounting for the incomplete and transitory efficacy of current anti-cancer therapies. However, neoplastic cells alone do not manifest the disease, but conscript a battery of non-tumor cells to enable and sustain hallmark capabilities of cancer. Escaping immunosurveillance is one of such capabilities. Tumors evolve immunosuppressive microenvironment to subvert anti-tumor immunity. In this review, we will focus on tumor-associated myeloid cells, which constitute an essential part of the immune microenvironment and reciprocally interact with cancer cells to establish malignancy toward metastasis. The diversity and plasticity of these cells constitute another layer of heterogeneity, beyond the heterogeneity of cancer cells themselves. We envision that immune microenvironment co-evolves with the genetic heterogeneity of tumor. Addressing the question of how genetically distinct tumors shape and are shaped by unique immune microenvironment will provide an attractive rationale to develop novel immunotherapeutic modalities. Here, we discuss the complex nature of tumor microenvironment, with an emphasis on the cellular and functional heterogeneity among tumor-associated myeloid cells as well as immune environment heterogeneity in the context of a full spectrum of human breast cancers.
Collapse
|
46
|
Padwal M, Siddique I, Wu L, Tang K, Boivin F, Liu L, Robertson J, Bridgewater D, West-Mays J, Gangji A, Brimble KS, Margetts PJ. Matrix metalloproteinase 9 is associated with peritoneal membrane solute transport and induces angiogenesis through β-catenin signaling. Nephrol Dial Transplant 2017; 32:50-61. [PMID: 27190383 DOI: 10.1093/ndt/gfw076] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 03/16/2016] [Indexed: 12/21/2022] Open
Abstract
Background For patients using peritoneal dialysis (PD), the peritoneal membrane can develop fibrosis and angiogenesis, leading to ultrafiltration failure, chronic hypervolemia and increased risk of technique failure and mortality. Matrix metalloproteinases (MMPs), and specifically the gelatinases (MMP2 and MMP9), may be involved in peritoneal membrane injury. Methods From stable PD patients, mesothelial cells were assayed for MMP gene expression. MMP9 was overexpressed in mouse peritoneum by adenovirus, and MMP9 -/- mice were subjected to transforming growth factor β (TGF-β)-induced peritoneal fibrosis. Results MMP9 mRNA expression correlated with peritoneal membrane solute transport properties. Overexpression of MMP9 in the mouse peritoneum induced submesothelial thickening and angiogenesis. MMP9 induced mesothelial cell transition to a myofibroblast phenotype measured by increased alpha smooth muscle actin and decreased E-cadherin expression. Angiogenesis was markedly reduced in MMP9 -/- mice treated with an adenovirus expressing active TGF-β compared with wild-type mice. TGF-β-mediated E-cadherin cleavage was MMP9 dependent, and E-cadherin cleavage led to β-catenin-mediated signaling. A β-catenin inhibitor blocked the angiogenic response induced by AdMMP9. Conclusions Our data suggest that MMP9 is involved in peritoneal membrane injury possibly through cleavage of E-cadherin and induction of β-catenin signaling. MMP9 is a potential biomarker for peritoneal membrane injury and is a therapeutic target to protect the peritoneal membrane in PD patients.
Collapse
Affiliation(s)
- Manreet Padwal
- Division of Nephrology, St. Joseph's Hospital, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Imad Siddique
- Division of Nephrology, St. Joseph's Hospital, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Lili Wu
- Institute of Traditional Chinese Medicine, Beijing, China
| | - Katelynn Tang
- Division of Nephrology, St. Joseph's Hospital, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Felix Boivin
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Limin Liu
- Division of Nephrology, St. Joseph's Hospital, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jennifer Robertson
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Darren Bridgewater
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Judith West-Mays
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Azim Gangji
- Division of Nephrology, St. Joseph's Hospital, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Kenneth Scott Brimble
- Division of Nephrology, St. Joseph's Hospital, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Peter J Margetts
- Division of Nephrology, St. Joseph's Hospital, Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
47
|
Dong W, Tang XL, Shang GB, Xu GL, Zhu WF, Liu HN. Dampness-Heat Accelerates DMBA-Induced Mammary Tumors in Rats. Chin J Integr Med 2017; 24:758-762. [DOI: 10.1007/s11655-017-2821-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2014] [Indexed: 10/18/2022]
|
48
|
Kim J, Mirando AC, Popel AS, Green JJ. Gene delivery nanoparticles to modulate angiogenesis. Adv Drug Deliv Rev 2017; 119:20-43. [PMID: 27913120 PMCID: PMC5449271 DOI: 10.1016/j.addr.2016.11.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 10/01/2016] [Accepted: 11/24/2016] [Indexed: 01/19/2023]
Abstract
Angiogenesis is naturally balanced by many pro- and anti-angiogenic factors while an imbalance of these factors leads to aberrant angiogenesis, which is closely associated with many diseases. Gene therapy has become a promising strategy for the treatment of such a disordered state through the introduction of exogenous nucleic acids that express or silence the target agents, thereby engineering neovascularization in both directions. Numerous non-viral gene delivery nanoparticles have been investigated towards this goal, but their clinical translation has been hampered by issues associated with safety, delivery efficiency, and therapeutic effect. This review summarizes key factors targeted for therapeutic angiogenesis and anti-angiogenesis gene therapy, non-viral nanoparticle-mediated approaches to gene delivery, and recent gene therapy applications in pre-clinical and clinical trials for ischemia, tissue regeneration, cancer, and wet age-related macular degeneration. Enhanced nanoparticle design strategies are also proposed to further improve the efficacy of gene delivery nanoparticles to modulate angiogenesis.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Adam C Mirando
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Departments of Ophthalmology, Neurosurgery, and Materials Science & Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
49
|
Chen XC, Wei XT, Guan JH, Shu H, Chen D. EGF stimulates glioblastoma metastasis by induction of matrix metalloproteinase-9 in an EGFR-dependent mechanism. Oncotarget 2017; 8:65969-65982. [PMID: 29029486 PMCID: PMC5630386 DOI: 10.18632/oncotarget.19622] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/30/2017] [Indexed: 01/24/2023] Open
Abstract
Epidermal growth factor (EGF) and EGF receptor (EGFR) play prominent roles in the metastasis of glioblastoma (GBM). However, the molecular mechanisms for the function of EGF and EGFR in GBM metastasis have not been elucidated. Herein, we demonstrate that coactivation of EGF and EGFR drives tumor metastasis in a matrix metalloproteinase-9 (MMP-9)-dependent manner. Expression levels of EGF, EGFR, and MMP-9 were substantially upregulated in the GBM and edema zones of patients, compared with those of paired unaffected participants. Secretion of EGF and MMP-9 was reduced in the cerebrospinal fluid (CSF) after removing GBM for 2 weeks by operation. To the mechanism, MMP-9 was upregulated by activating EGF and EGFR via PI3K/AKT- and ERK1/2-dependent pathways. Moreover, signal transducer and activator of transcription (STAT) 3 and STAT5 mediated the activation of NF-κB by PI3K/AKT and ERK1/2 pathways. This resulted in transactivation of MMP-9 in GBM. Finally, MMP-9 induction facilitated abnormal proliferation, migration, and invasion of cells, which contributed to GBM metastasis.
Collapse
Affiliation(s)
- Xing-Chen Chen
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, P. R. China`
| | - Xiang-Tai Wei
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, P. R. China`
| | - Jun-Hong Guan
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, P. R. China`
| | - Hong Shu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, P. R. China`
| | - Duo Chen
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, P. R. China`
| |
Collapse
|
50
|
Ward MP, Spiers JP. Protein phosphatase 2A regulation of markers of extracellular matrix remodelling in hepatocellular carcinoma cells: functional consequences for tumour invasion. Br J Pharmacol 2017; 174:1116-1130. [PMID: 28239848 DOI: 10.1111/bph.13759] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 02/13/2017] [Accepted: 02/16/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE A hallmark of tumour invasion is breakdown of the extracellular matrix due to dysregulation of the matrix metalloproteinase (MMP) system. While our understanding of how this is regulated by kinase signalling pathways is well established, its counter-regulation by protein phosphatases (PP) is poorly understood. Therefore, we investigated the effect of PP inhibition on markers of extracellular remodelling and how PP2A activity modulated MMP-9 abundance and function of Hep3B cells. EXPERIMENTAL APPROACH Cells were exposed to okadaic acid (OA), tautomycetin and cyclosporin A, and the expression profile determined using PCR. Effects of OA and a protein inhibitor of PP2A, CIP2A, on MMP-9 abundance, PP2A activity and cell migration were investigated using ELISA, promoter constructs, siRNA knockdown and transwell migration assays. KEY RESULTS OA increased expression and abundance of MMP-9 and the tissue inhibitor of MMP, TIMP-1, without affecting other MMPs, TIMPs and ADAMs. The effect on MMP-9 was mimicked by CIP2A overexpression and knockdown of the PPP2CA catalytic, but not PPP2R1A scaffolding, subunit. Cyclosporin A and PPP1CA silencing did not alter MMP-9 expression, while tautomycetin transiently increased it. Mutation of AP-1, but not NF-κB, binding sites inhibited OA-mediated MMP-9 transcriptional activity. OA and CIP2A decreased PP2A activity and increased cell migration. CONCLUSION AND IMPLICATIONS OA increased MMP-9 by decreasing PP2A activity and PP2Ac, through AP-1 binding sites on the MMP-9 promoter. The functional consequence of this and CIP2A overexpression was increased cell migration. Hence, PP2A inhibition induced a metastatic phenotype through alterations in MMP-9 in Hep3B cells.
Collapse
Affiliation(s)
- M P Ward
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| | - J P Spiers
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|