1
|
Todorović S, Ćeranić MS, Tošković B, Diklić M, Mitrović Ajtić O, Subotički T, Vukotić M, Dragojević T, Živković E, Oprić S, Stojiljkovic M, Gačić J, Čolaković N, Crnokrak B, Čokić VP, Đikić D. Proinflammatory Microenvironment in Adenocarcinoma Tissue of Colorectal Carcinoma. Int J Mol Sci 2024; 25:10062. [PMID: 39337548 PMCID: PMC11432548 DOI: 10.3390/ijms251810062] [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: 08/19/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer-promoting proinflammatory microenvironment influences colorectal cancer (CRC) development. We examined the biomarkers of inflammation, intestinal differentiation, and DNA activity correlated with the clinical parameters to observe progression and prognosis in the adenocarcinoma subtype of CRC. Their immunohistology, immunoblotting, and RT-PCR analyses were performed in the adenocarcinoma and neighboring healthy tissues of 64 patients with CRC after routine colorectal surgery. Proinflammatory nuclear factor kappa B (NFκB) signaling as well as interleukin 6 (IL-6) and S100 protein levels were upregulated in adenocarcinoma compared with nearby healthy colon tissue. In contrast to nitrotyrosine expression, the oxidative stress marker 8-Hydroxy-2'-deoxyguanosine (8-OHdG) was increased in adenocarcinoma tissue. Biomarkers of intestinal differentiation β-catenin and mucin 2 (MUC2) were inversely regulated, with the former upregulated in adenocarcinoma tissue and positively correlated with tumor marker CA19-9. Downregulation of MUC2 expression correlated with the increased 2-year survival rate of patients with CRC. Proliferation-related mammalian target of rapamycin (mTOR) signaling was activated, and Ki67 frequency was three-fold augmented in positive correlation with metastasis and cancer stage, respectively. Conclusion: We demonstrated a parallel induction of oxidative stress and inflammation biomarkers in adenocarcinoma tissue that was not reflected in the neighboring healthy colon tissue of CRC. The expansiveness of colorectal adenocarcinoma was confirmed by irregular intestinal differentiation and elevated proliferation biomarkers, predominantly Ki67. The origin of the linked inflammatory factors was in adenocarcinoma tissue, with an accompanying systemic immune response.
Collapse
Affiliation(s)
- Slobodan Todorović
- University Hospital Medical Center Bežanijska Kosa, Faculty of Medicine, University of Belgrade, Dr. Žorža Matea bb, 11080 Belgrade, Serbia
| | - Miljan S Ćeranić
- University Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Pasterova 2, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr. Subotića starijeg 8, 11000 Belgrade, Serbia
| | - Borislav Tošković
- University Hospital Medical Center Bežanijska Kosa, Faculty of Medicine, University of Belgrade, Dr. Žorža Matea bb, 11080 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr. Subotića starijeg 8, 11000 Belgrade, Serbia
| | - Miloš Diklić
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, 11129 Belgrade, Serbia
| | - Olivera Mitrović Ajtić
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, 11129 Belgrade, Serbia
| | - Tijana Subotički
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, 11129 Belgrade, Serbia
| | - Milica Vukotić
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, 11129 Belgrade, Serbia
| | - Teodora Dragojević
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, 11129 Belgrade, Serbia
| | - Emilija Živković
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, 11129 Belgrade, Serbia
| | - Svetlana Oprić
- University Hospital Medical Center Bežanijska Kosa, Faculty of Medicine, University of Belgrade, Dr. Žorža Matea bb, 11080 Belgrade, Serbia
| | - Miodrag Stojiljkovic
- University Hospital Medical Center Bežanijska Kosa, Faculty of Medicine, University of Belgrade, Dr. Žorža Matea bb, 11080 Belgrade, Serbia
| | - Jasna Gačić
- University Hospital Medical Center Bežanijska Kosa, Faculty of Medicine, University of Belgrade, Dr. Žorža Matea bb, 11080 Belgrade, Serbia
| | - Nataša Čolaković
- University Hospital Medical Center Bežanijska Kosa, Faculty of Medicine, University of Belgrade, Dr. Žorža Matea bb, 11080 Belgrade, Serbia
| | - Bogdan Crnokrak
- University Hospital Medical Center Bežanijska Kosa, Faculty of Medicine, University of Belgrade, Dr. Žorža Matea bb, 11080 Belgrade, Serbia
| | - Vladan P Čokić
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, 11129 Belgrade, Serbia
| | - Dragoslava Đikić
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, 11129 Belgrade, Serbia
| |
Collapse
|
2
|
Hoang SH, Dao H, Lam EM. A network pharmacology approach to elucidate the anti-inflammatory effects of ellagic acid. J Biomol Struct Dyn 2024; 42:7409-7420. [PMID: 37522847 DOI: 10.1080/07391102.2023.2240417] [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: 01/11/2023] [Accepted: 07/17/2023] [Indexed: 08/01/2023]
Abstract
Ellagic acid (EA) is a naturally occurring polyphenolic compound found in various fruits and vegetables like strawberries, raspberries, pomegranates, and nuts such as pecans and walnuts. With its antioxidant properties, EA has shown potential health benefits, although further research is necessary to fully comprehend its effects, mechanisms, and safe and effective application as a complementary medicine. Notably, there is accumulating evidence of EA's anti-inflammatory effects; however, the precise underlying mechanism remains unclear. To investigate the anti-inflammatory properties of EA, a network pharmacology approach was employed. The study identified 52 inflammation-related targets of EA and revealed significant signaling pathways and relevant diseases associated with inflammation through GO and KEGG analysis. Furthermore, topological analysis identified 10 important targets, including AKT1, VEGFA, TNF, MAPK3, ALB, SELP, MMP9, MMP2, PTGS2, and ICAM1. Molecular docking and molecular dynamics simulations were conducted, indicating that AKT1, PTGS2, VEGFA, and MAPK3 are the most likely targets of EA, as evidenced by their molecular mechanics Poisson-Boltzmann surface area binding energy calculations. In summary, this study not only confirmed the anti-inflammatory effects of EA observed in previous research but also identified the most probable targets of EA.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Skyler H Hoang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| | - Hue Dao
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| | - Emerson My Lam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| |
Collapse
|
3
|
Abdelaziz I, Bounaama A, Djerdjouri B, Amir-Tidadini ZC. Low-dose dimethylfumarate attenuates colitis-associated cancer in mice through M2 macrophage polarization and blocking oxidative stress. Toxicol Appl Pharmacol 2024; 489:117018. [PMID: 38945373 DOI: 10.1016/j.taap.2024.117018] [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: 02/01/2024] [Revised: 06/13/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
Colitis-associated cancer (CAC) is an aggressive subtype of colorectal cancer that can develop in ulcerative colitis patients and is driven by chronic inflammation and oxidative stress. Current chemotherapy for CAC, based on 5-fluorouracil and oxalipltin, is not fully effective and displays severe side effects, prompting the search for alternative therapies. Dimethylfumarate (DMF), an activator of the nuclear factor erythroid 2-related factor 2 (NRF2), is a potent antioxidant and immunomodelatrory drug used in the treatment of multiple sclerosis and showed a strong anti-inflammatory effect on experimental colitis. Here, we investigated the chemotherapeutic effect of DMF on an experimental model of CAC. Male NMRI mice were given two subcutaneous injections of 1,2 Dimethylhydrazine (DMH), followed by three cycles of dextran sulfate sodium (DSS). Low-dose (DMF30) and high-dose of DMF (DMF100) or oxaliplatin (OXA) were administered from the 8th to 12th week of the experiment, and then the colon tissues were analysed histologically and biochemically. DMH/DSS induced dysplastic aberrant crypt foci (ACF), oxidative stress, and severe colonic inflammation, with a predominance of pro-inflammatory M1 macrophages. As OXA, DMF30 reduced ACF multiplicity and crypt dysplasia, but further restored redox status, and reduced colitis severity by shifting macrophages towards the anti-inflammatory M2 phenotype. Surprisingly, DMF100 exacerbated ACF multiplicity, oxidative stress, and colon inflammation, likely through NRF2 and p53 overexpression in colonic inflammatory cells. DMF had a dual effect on CAC. At low dose, DMF is chemotherapeutic and acts as an antioxidant and immunomodulator, whereas at high dose, DMF is pro-oxidant and exacerbates colitis-associated cancer.
Collapse
Affiliation(s)
- Ismahane Abdelaziz
- Tamayouz_Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Algiers, Algeria
| | - Abdelkader Bounaama
- Tamayouz_Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Algiers, Algeria.
| | - Bahia Djerdjouri
- Tamayouz_Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Algiers, Algeria
| | | |
Collapse
|
4
|
Ma L, Wu Y, Luo J, Li F, Zhang M, Cai Y, Dai Y, Pi Z, Zheng F, Yue H. Identifying the active compounds and mechanism of action of TongFu XieXia Decoction for treating intestinal obstruction using network pharmacology combined with ultra-high performance liquid chromatography-quadrupole-orbitrap mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2024; 38:e9664. [PMID: 38124169 DOI: 10.1002/rcm.9664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 12/23/2023]
Abstract
RATIONALE TongFu XieXia Decoction (TFXXD), a formulation rooted in traditional Chinese medicine and optimized through clinical practice, serves as an advanced version of the classic Da Cheng Qi decoction used for treating intestinal obstruction (IO), demonstrating significant therapeutic efficacy. However, due to the intricate nature of herbal compositions, the principal constituents and potential mechanisms of TFXXD have yet to be clarified. Accordingly, this study seeks to identify the active compounds and molecular targets of TFXXD, as well as to elucidate its anti-IO mechanisms. METHODS Qualitative identification of the principal constituents of TFXXD was accomplished using ultra-high preformance liquid chromatography-quadrupole-orbitrap mass spectrometry (UPLC-Q-Orbitrap-MS/MS) analysis. PharmMapper facilitated the prediction of potential molecular targets, whereas protein-protein interaction analysis was conducted using STRING 11.0. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed using the Metascape database. A "compounds-target-pathway" network was meticulously constructed within Cytoscape 3.8.2. Finally, molecular docking studies were performed to investigate the interactions between the core target and the crucial compound. RESULTS UPLC-Q-Orbitrap-MS/MS analysis identified 65 components with high precision and sensitivity. Furthermore, 64 potential targets were identified as integral to TFXXD bioactivity in IO treatment. Gene Ontology enrichment analysis revealed 995 distinct biological functions, while the Kyoto Encyclopedia of Genes and Genomes enrichment analysis identified 143 intricate signaling pathways. CONCLUSION Molecular docking studies substantiated the substantial affinity between the TFXXD bioactive constituents and their corresponding targets in the context of IO. TFXXD exerts its therapeutic efficacy in IO through a multifaceted interplay between multiple compounds, targets, and pathways. The integration of network pharmacology with UPLC-Q-Orbitrap-MS/MS has emerged as a promising strategy to unravel the intricate web of molecular interactions underlying herbal medicine. However, it is imperative to emphasize the necessity for further in vivo and in vitro experiments.
Collapse
Affiliation(s)
- Liting Ma
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yongxi Wu
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jing Luo
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Fangtong Li
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Meiyu Zhang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yongyu Cai
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yulin Dai
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zifeng Pi
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Fei Zheng
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hao Yue
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
5
|
Dutta RK, Abu YF, Tao J, Chupikova I, Oleas J, Singh PK, Vitari NA, Qureshi R, Ramakrishnan S, Roy S. Altered gut microbiome drives heightened pain sensitivity in a murine model of metastatic triple-negative breast cancer. Am J Cancer Res 2024; 14:274-299. [PMID: 38323292 PMCID: PMC10839306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/25/2023] [Indexed: 02/08/2024] Open
Abstract
The microbiota residing in the gut environment is essential for host homeostasis. Increasing evidence suggests that microbial perturbation (dysbiosis) regulates cancer initiation and progression at local and distant sites. Here, we have identified microbial dysbiosis with the depletion of commensal bacteria as a host-intrinsic factor associated with metastatic dissemination to the bone. Using a mouse model of triple-negative mammary cancer, we demonstrate that a pre-established disruption of microbial homeostasis using an antibiotic cocktail increases tumor growth, enhanced circulating tumor cells, and subsequent dissemination to the bone. We found that the presence of pathogenic bacteria and loss of commensal bacteria in an antibiotic-induced gut environment is associated with sustained inflammation. Increased secretion of G-CSF and MMP-9 in intestinal tissues, followed by increased neutrophil infiltration and severe systemic inflammation in tumor-bearing mice, indicates the direct consequence of a dysbiotic microbiome. Increased neutrophil infiltration to the bone metastatic niche facilitates extravasation and transendothelial migration of tumor cells. It provides a novel, pre-established, and favorable environment to form an immunosuppressive pre-metastatic niche. The presence of tumor cells in immunosuppressive metastatic tumor niche disrupts the balance between osteoblasts and osteoclasts, promotes osteoclast differentiation, and remodels the bone structure. Excessive bone resorption by osteoclasts causes bone degradation and ultimately causes extreme pain in a bone metastatic mouse model. In clinical settings, bone metastasis is associated with intractable severe pain that severely compromises the quality of life in these patients.
Collapse
Affiliation(s)
- Rajib K Dutta
- Department of Surgery, University of MiamiMiami, FL 33136, USA
| | - Yaa F Abu
- Department of Surgery, University of MiamiMiami, FL 33136, USA
- Department of Microbiology and Immunology, University of MiamiMiami, FL 33136, USA
| | - Junyi Tao
- Department of Surgery, University of MiamiMiami, FL 33136, USA
| | - Irina Chupikova
- Department of Surgery, University of MiamiMiami, FL 33136, USA
| | - Janneth Oleas
- Department of Surgery, University of MiamiMiami, FL 33136, USA
| | - Praveen K Singh
- Department of Surgery, University of MiamiMiami, FL 33136, USA
| | - Nicolas A Vitari
- Department of Surgery, University of MiamiMiami, FL 33136, USA
- Department of Microbiology and Immunology, University of MiamiMiami, FL 33136, USA
| | - Rehana Qureshi
- Department of Pathology, University of MiamiMiami, FL 33136, USA
| | | | - Sabita Roy
- Department of Surgery, University of MiamiMiami, FL 33136, USA
| |
Collapse
|
6
|
Yang JH, Park JW, Kim HS, Lee S, Yerke AM, Jaiswal YS, Williams LL, Hwang S, Moon KH. Effects of Antibiotic Residues on Fish Gut Microbiome Dysbiosis and Mucosal Barrier-Related Pathogen Susceptibility in Zebrafish Experimental Model. Antibiotics (Basel) 2024; 13:82. [PMID: 38247641 PMCID: PMC10812462 DOI: 10.3390/antibiotics13010082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/05/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
The symbiotic community of microorganisms in the gut plays an important role in the health of the host. While many previous studies have been performed on the interactions between the gut microbiome and the host in mammals, studies in fish are still lacking. In this study, we investigated changes in the intestinal microbiome and pathogen susceptibility of zebrafish (Danio rerio) following chronic antibiotics exposure. The chronic antibiotics exposure assay was performed on zebrafish for 30 days using oxytetracycline (Otc), sulfamethoxazole/trimethoprim (Smx/Tmp), or erythromycin (Ery), which are antibiotics widely used in the aquaculture industry. The microbiome analysis indicated that Fusobacteria, Proteobacteria, Firmicutes, and Bacteroidetes were the dominant phyla in the gut microbiome of the zebrafish used in this study. However, in Smx/Tmp-treated zebrafish, the compositions of Fusobacteria and Proteobacteria were changed significantly, and in Ery-treated zebrafish, the compositions of Proteobacteria and Firmicutes were altered significantly. Although alpha diversity analysis showed that there was no significant difference in the richness, beta diversity analysis revealed a community imbalance in the gut microbiome of all chronically antibiotics-exposed zebrafish. Intriguingly, in zebrafish with dysbiosis in the gut microbiome, the pathogen susceptibility to Edwardsiella piscicida, a representative Gram-negative fish pathogen, was reduced. Gut microbiome imbalance resulted in a higher count of goblet cells in intestinal tissue and an upregulation of genes related to the intestinal mucosal barrier. In addition, as innate immunity was enhanced by the increased mucosal barrier, immune and stress-related gene expression in the intestinal tissue was downregulated. In this study, we provide new insight into the effect of gut microbiome dysbiosis on pathogen susceptibility.
Collapse
Affiliation(s)
- Jun Hyeok Yang
- Laboratory of Marine Microbiology, Division of Convergence of Marine Science, Korea Maritime & Ocean University, Busan 49112, Republic of Korea; (J.H.Y.); (J.W.P.); (H.S.K.)
- Department of Marine Bioscience and Environment, Korea Maritime & Ocean University, Busan 49112, Republic of Korea
| | - Jeong Woo Park
- Laboratory of Marine Microbiology, Division of Convergence of Marine Science, Korea Maritime & Ocean University, Busan 49112, Republic of Korea; (J.H.Y.); (J.W.P.); (H.S.K.)
- Department of Convergence Study on the Ocean Science and Technology, Ocean Science and Technology School, Korea Maritime & Ocean University, Busan 49112, Republic of Korea
| | - Ho Sung Kim
- Laboratory of Marine Microbiology, Division of Convergence of Marine Science, Korea Maritime & Ocean University, Busan 49112, Republic of Korea; (J.H.Y.); (J.W.P.); (H.S.K.)
- Department of Convergence Study on the Ocean Science and Technology, Ocean Science and Technology School, Korea Maritime & Ocean University, Busan 49112, Republic of Korea
| | - Seungki Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Republic of Korea;
| | - Aaron M. Yerke
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA;
| | - Yogini S. Jaiswal
- Center for Excellence in Post Harvest Technologies, North Carolina Agricultural and Technical State University, The North Carolina Research Campus, 500 Laureate Way, Kannapolis, NC 28081, USA; (Y.S.J.); (L.L.W.)
| | - Leonard L. Williams
- Center for Excellence in Post Harvest Technologies, North Carolina Agricultural and Technical State University, The North Carolina Research Campus, 500 Laureate Way, Kannapolis, NC 28081, USA; (Y.S.J.); (L.L.W.)
| | - Sungmin Hwang
- Division of Practical Research, Honam National Institute Biological Resources, Mokpo-si 58762, Republic of Korea
| | - Ki Hwan Moon
- Laboratory of Marine Microbiology, Division of Convergence of Marine Science, Korea Maritime & Ocean University, Busan 49112, Republic of Korea; (J.H.Y.); (J.W.P.); (H.S.K.)
- Department of Marine Bioscience and Environment, Korea Maritime & Ocean University, Busan 49112, Republic of Korea
- Department of Convergence Study on the Ocean Science and Technology, Ocean Science and Technology School, Korea Maritime & Ocean University, Busan 49112, Republic of Korea
| |
Collapse
|
7
|
Tufail M. Unlocking the potential of the tumor microenvironment for cancer therapy. Pathol Res Pract 2023; 251:154846. [PMID: 37837860 DOI: 10.1016/j.prp.2023.154846] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/15/2023] [Accepted: 10/02/2023] [Indexed: 10/16/2023]
Abstract
The tumor microenvironment (TME) holds a crucial role in the progression of cancer. Epithelial-derived tumors share common traits in shaping the TME. The Warburg effect is a notable phenomenon wherein tumor cells exhibit resistance to apoptosis and an increased reliance on anaerobic glycolysis for energy production. Recognizing the pivotal role of the TME in controlling tumor growth and influencing responses to chemotherapy, researchers have focused on developing potential cancer treatment strategies. A wide array of therapies, including immunotherapies, antiangiogenic agents, interventions targeting cancer-associated fibroblasts (CAF), and therapies directed at the extracellular matrix, have been under investigation and have demonstrated efficacy. Additionally, innovative techniques such as tumor tissue explants, "tumor-on-a-chip" models, and multicellular tumor spheres have been explored in laboratory research. This comprehensive review aims to provide insights into the intricate cross-talk between cancer-associated signaling pathways and the TME in cancer progression, current therapeutic approaches targeting the TME, the immune landscape within solid tumors, the role of the viral TME, and cancer cell metabolism.
Collapse
Affiliation(s)
- Muhammad Tufail
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
8
|
Seth P, Dubey S. IL-22 as a target for therapeutic intervention: Current knowledge on its role in various diseases. Cytokine 2023; 169:156293. [PMID: 37441942 DOI: 10.1016/j.cyto.2023.156293] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/12/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023]
Abstract
IL-22 has emerged as a crucial cytokine mediating protective response against pathogens and tissue regeneration. Dysregulated production of IL-22 has been shown to play a pivotal role in the pathogenesis of various diseases like malignant tumours, viral, cardiovascular, allergic and autoimmune disorders. Interleukin 22 belongs to IFN-IL-10 cytokine family. It is a major proinflammatory cytokine secreted by activated Th1 cells (Th22), though can also be secreted by many other immune cells like group 3 innate lymphocytes, γδ T cells, NK cells, NK T cells, and mucosal associated invariant T cells. Th22 cells exclusively release IL-22 but not IL-17 or IFN-γ (as Th1 cells releases IFN-γ along with IL-22 and Th17 cells releases IL-17 along with IL-22) and also express aryl hydrocarbon receptor as the key transcription factor. Th22 cells also exhibit expression of chemokine receptor CCR6 and skin-homing receptors CCR4 and CCR10 indicating the involvement of this subset in bolstering epithelial barrier immunity and promoting secretion of antimicrobial peptides (AMPs) from intestinal epithelial cells. The function of IL-22 is modulated by IL-22 binding protein (binds to IL-22 and inhibits it binding to its cell surface receptor); which serves as a competitor for IL-22R1 chain of IL-22 receptor. The pathogenic and protective nature of the Th22 cells is modulated both by the site of infected tissue and the type of disease pathology. This review aims to discuss key features of IL-22 biology, comparisons between IL and 22 and IFN-γ and its role as a potential immune therapy target in different maladies.
Collapse
Affiliation(s)
- Pranav Seth
- Amity Institute of Virology & Immunology, Amity University Uttar Pradesh, Sector 125, Noida, India
| | - Shweta Dubey
- Amity Institute of Virology & Immunology, Amity University Uttar Pradesh, Sector 125, Noida, India.
| |
Collapse
|
9
|
Kyrian O, Dorofeyev A, Derkach I, Zhigal Y. Evaluation of the Impact of Some Single-Nucleotide Gene Polymorphisms on the Development of Adenomatous Polyps of the Colon in Patients with Irritable Bowel Syndrome. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2023; 34:822-830. [PMID: 37404119 PMCID: PMC10544543 DOI: 10.5152/tjg.2023.21985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 10/09/2022] [Indexed: 07/06/2023]
Abstract
BACKGROUND The number of cases of irritable bowel syndrome is growing worldwide, in which adenomatous polyps can develop as a result of microinflammation of the colonic epithelium. Our study was aimed at the identification of the possible effect of single-nucleotide polymorphisms on the risk of the development of irritable bowel syndrome-related colonic adenomatous polyps. MATERIALS AND METHODS The study involved 187 irritable bowel syndrome patients. The single-nucleotide polymorphisms were investigated by the polymerase chain reaction method and DNA was extracted with the phenol-chloroform: interleukin-1β gene-31C/T (rs1143627), -511C/T (rs16944); interleukin-6 gene-174G/C (rs1800795); interleukin-10 gene-592C/A (rs1800872), -819T/C (rs1800871), -1082A/G (rs1800896); Toll-like receptor-2 gene Arg753Gln (rs5743708); Toll-like receptor-4 gene Thr399ile (rs4986791), Asp299Gly (rs4986790); and metalloproteinase-9 gene-8202A/G (rs11697325). The study of polymorphic loci was checked for compliance with the Hardy- Weinberg equilibrium using Fisher's exact test along with the analyses of the frequency of alleles and the genotypes. RESULTS The association of diseases with G allele Toll-like receptor-2 gene Arg753Gln (rs5743708) was revealed in irritable bowel syndrome patients with adenomatous polyps of the colon (P < .0006) and AG single-nucleotide polymorphisms s of Toll-like receptor-2 gene (χ2 = 12.78, P < .002); A allele had a protective effect. The AG genotype metalloproteinase-9 gene-8202A/G (rs11697325) polymorphism in irritable bowel syndrome patients with adenomatous polyps of the colon had a protective effect (P < .05). AA genotype interleukin-10 gene-1082A/G (rs1800896) polymorphism in the irritable bowel syndrome patient (χ2 = 33.97, 4.0E-8) can be considered as the risk for adenomatous polyps of the colon in irritable bowel syndrome. CONCLUSION G allele Toll-like receptor-2 gene Arg753Gln (rs5743708) and AA genotype interleukin-10 gene-1082A/G (rs1800896) polymorphisms can be the marker of the emergence of adenomatous polyps of the colon concomitant with irritable bowel syndrome.
Collapse
Affiliation(s)
- Olena Kyrian
- Department of Family Medicine and Therapy, Poltava State Medical University, Poltava, Ukraine
| | - Andrey Dorofeyev
- Chair of Internal Medicine and Gerontology, Shupyk National University of Public Health of Ukraine, Kyiv, Ukraine
| | - Igor Derkach
- Department of Urology, Truskavets City Hospital, Truskavets, Ukraine
| | - Yulia Zhigal
- Chair of Internal Medicine and Gerontology, Shupyk National University of Public Health of Ukraine, Kyiv, Ukraine
| |
Collapse
|
10
|
Kumari S, Kumar P. Design and Computational Analysis of an MMP9 Inhibitor in Hypoxia-Induced Glioblastoma Multiforme. ACS OMEGA 2023; 8:10565-10590. [PMID: 36969457 PMCID: PMC10035023 DOI: 10.1021/acsomega.3c00441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The main therapeutic difficulties in treating hypoxia-induced glioblastoma multiforme (GBM) are toxicity of current treatments and the resistance brought on by the microenvironment. More effective therapeutic alternatives are urgently needed to reduce tumor lethality. Hence, we screened plant-based natural product panels intending to identify novel drugs without elevating drug resistance. We explored GEO for the hypoxia GBM model and compared hypoxic genes to non-neoplastic brain cells. A total of 2429 differentially expressed genes expressed exclusively in hypoxia were identified. The functional enrichment analysis demonstrated genes associated with GBM, further PPI network was constructed, and biological pathways associated with them were explored. Seven webtools, including GEPIA2.0, TIMER2.0, TCGA-GBM, and GlioVis, were used to validate 32 hub genes discovered using Cytoscape tool in GBM patient samples. Four GBM-specific hypoxic hub genes, LYN, MMP9, PSMB9, and TIMP1, were connected to the tumor microenvironment using TIMER analysis. 11 promising hits demonstrated positive drug-likeness with nontoxic characteristics and successfully crossed blood-brain barrier and ADMET analyses. Top-ranking hits have stable intermolecular interactions with the MMP9 protein according to molecular docking, MD simulation, MM-PBSA, PCA, and DCCM analyses. Herein, we have reported flavonoids, 7,4'-dihydroxyflavan, (3R)-3-(4-hydroxybenzyl)-6-hydroxy-8-methoxy-3,4-dihydro-2H-1-benzopyran, and 4'-hydroxy-7-methoxyflavan, to inhibit MMP9, a novel hypoxia gene signature that could serve as a promising predictor in various clinical applications, including GBM diagnosis, prognosis, and targeted therapy.
Collapse
|
11
|
Zheng S, Zhang D, Duan B, Mo G, Li J, Huang H, Wang S, Ye Y, Huang Z, Huang P, Zhang F, Huang F, Han L. Metabolomics integrated network pharmacology reveals the mechanism of Ma-Mu-Ran Antidiarrheal Capsules on acute enteritis mice. Anal Biochem 2023; 668:115116. [PMID: 36925055 DOI: 10.1016/j.ab.2023.115116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Acute enteritis (AE) is a type of digestive disease caused by biochemical factors that irritate the intestinal tract or pathogenic bacteria that infect it. In China, Ma-Mu-Ran Antidiarrheal Capsules (MMRAC) have been applied against diarrhea caused by AE and bacillary dysentery for many years, but the underlying mechanisms of their beneficial effects are not known. In the present study, network pharmacology and metabolomics were performed to clarify the active ingredients of MMRAC and explore the specific mechanism of MMRAC on AE mice. A total of 43 active components of MMRAC with 87 anti-AE target genes were identified, and these target genes were enriched in IL-17 and HIF-1 signaling pathways. Integration analysis revealed that purine metabolism was the critical metabolic pathway by which MMRAC exerted its therapeutic effect against AE. Specifically, MAPK14, MMP9, PTGS2, HIF1A, EGLN1, NOS2 were the pivotal targets of MMRAC for the treatment of AE, and Western blot analysis revealed MMRAC to decrease protein levels of these pro-inflammatory signaling molecules. According to molecular docking, these key targets have a strong affinity with the MMRAC compounds. Collectively, MMRAC relieved the colon inflammation of AE mice via regulating inflammatory signaling pathways to reduce hypoxia and improved energy metabolism.
Collapse
Affiliation(s)
- Sili Zheng
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Dongning Zhang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China; Xinjiang Uygur Pharmaceutical Co., LTD, Urumqi, Xinjiang, 830026, China
| | - Bailu Duan
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Guoyan Mo
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China; Key Laboratory of Traditional Chinese Medicine Resource and Prescription, Ministry of Education, Wuhan, Hubei, 430065, China
| | - Jingjing Li
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Hailing Huang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Shanshan Wang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Yan Ye
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Zhuang Huang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Ping Huang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Fengyun Zhang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Fang Huang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China.
| | - Lintao Han
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China; Key Laboratory of Traditional Chinese Medicine Resource and Prescription, Ministry of Education, Wuhan, Hubei, 430065, China.
| |
Collapse
|
12
|
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
|
13
|
Reiss Z, Rob F, Kolar M, Schierova D, Kreisinger J, Jackova Z, Roubalova R, Coufal S, Mihula M, Thon T, Bajer L, Novakova M, Vasatko M, Kostovcikova K, Galanova N, Lukas M, Kverka M, Tresnak Hercogova J, Tlaskalova-Hogenova H, Jiraskova Zakostelska Z. Skin microbiota signature distinguishes IBD patients and reflects skin adverse events during anti-TNF therapy. Front Cell Infect Microbiol 2023; 12:1064537. [PMID: 36704107 PMCID: PMC9872723 DOI: 10.3389/fcimb.2022.1064537] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/01/2022] [Indexed: 01/11/2023] Open
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are two forms of inflammatory bowel disease (IBD), where the role of gut but not skin dysbiosis is well recognized. Inhibitors of TNF have been successful in IBD treatment, but up to a quarter of patients suffer from unpredictable skin adverse events (SkAE). For this purpose, we analyzed temporal dynamics of skin microbiota and serum markers of inflammation and epithelial barrier integrity during anti-TNF therapy and SkAE manifestation in IBD patients. We observed that the skin microbiota signature of IBD patients differs markedly from healthy subjects. In particular, the skin microbiota of CD patients differs significantly from that of UC patients and healthy subjects, mainly in the retroauricular crease. In addition, we showed that anti-TNF-related SkAE are associated with specific shifts in skin microbiota profile and with a decrease in serum levels of L-FABP and I-FABP in IBD patients. For the first time, we showed that shifts in microbial composition in IBD patients are not limited to the gut and that skin microbiota and serum markers of the epithelium barrier may be suitable markers of SkAE during anti-TNF therapy.
Collapse
Affiliation(s)
- Zuzana Reiss
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Filip Rob
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University, University Hospital Bulovka, Prague, Czechia
| | - Martin Kolar
- IBD Clinical and Research Centre ISCARE a.s., Prague, Czechia
| | - Dagmar Schierova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jakub Kreisinger
- Department of Zoology, Faculty of Science, Charles University, Prague, Czechia
| | - Zuzana Jackova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Radka Roubalova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Stepan Coufal
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Mihula
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Tomas Thon
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Lukas Bajer
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia,Department of Gastroenterology and Hepatology, Institute of Clinical and Experimental Medicine, Prague, Czechia
| | - Michaela Novakova
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University, University Hospital Bulovka, Prague, Czechia
| | - Martin Vasatko
- IBD Clinical and Research Centre ISCARE a.s., Prague, Czechia
| | - Klara Kostovcikova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Natalie Galanova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Milan Lukas
- IBD Clinical and Research Centre ISCARE a.s., Prague, Czechia,Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czechia
| | - Miloslav Kverka
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jana Tresnak Hercogova
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University, University Hospital Bulovka, Prague, Czechia,Prof. Hercogova Dermatology, Prague, Czechia
| | | | - Zuzana Jiraskova Zakostelska
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia,*Correspondence: Zuzana Jiraskova Zakostelska,
| |
Collapse
|
14
|
Kyrian OA. RISK FACTORS FOR NEOPLASIA IN PATIENS WITH ULCERATIVE COLITIS. BULLETIN OF PROBLEMS BIOLOGY AND MEDICINE 2023. [DOI: 10.29254/2077-4214-2022-4-167-167-178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
15
|
Zhou R, Guo T, Li J. Research progress on the antitumor effects of astragaloside IV. Eur J Pharmacol 2022; 938:175449. [PMID: 36473596 DOI: 10.1016/j.ejphar.2022.175449] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/15/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
One of the most important and effective components of Astragalus membranaceus is astragaloside IV (AS-IV), which can exert anti-tumor effects through various pathways. For instance, AS-IV exerts an anti-tumor effect by acting at the cellular level, regulating the phenotype switch of tumor-associated macrophages, or inhibiting the development of tumor cells. Furthermore, AS-IV inhibits tumor cell progression by enhancing its sensitivity to antitumor drugs or reversing the drug resistance of tumor cells. This article reviews the different mechanisms of AS-IV inhibition of epithelial-mesenchymal transition (EMT), migration, proliferation, and invasion of tumor cells, inducing apoptosis and improving the sensitivity of anti-tumor drugs. This review summarizes recent progress in the current research into AS-IV anti-tumor effect and provides insight on the next anti-tumor research of AS-IV.
Collapse
Affiliation(s)
- Ruixi Zhou
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou 730030, China
| | - Tiankang Guo
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730030, China
| | - Junliang Li
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730030, China; The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou 730030, China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730030, China.
| |
Collapse
|
16
|
Le C, Zeffren N, Kramer N, Rosenstein ED. Rheumatologic Associations of Microscopic Colitis: A Narrative Review. Mod Rheumatol 2022; 33:441-447. [PMID: 35993773 DOI: 10.1093/mr/roac080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/22/2022] [Accepted: 07/31/2022] [Indexed: 11/12/2022]
Abstract
Extra-intestinal manifestations are frequent complications of the classical inflammatory bowel diseases, Crohn's disease and ulcerative colitis. However, in addition to the classical diseases, there is a spectrum of conditions, often termed "microscopic colitis", in which extra-intestinal manifestations are less well described. Our objective was to review the literature regarding the extra-intestinal manifestations complicating microscopic colitis and describe the association with systemic autoimmune rheumatic diseases. A comprehensive search and review of peer-reviewed English-language and international journals and reports was completed based on key terms, including "microscopic colitis", "lymphocytic colitis", "collagenous colitis", "inflammatory bowel disease", "extraintestinal manifestations", and the specific disease associations utilizing the PubMed Central database and MEDLINE. A broad spectrum of rheumatologic manifestations has been reported in patients with microscopic colitis. The identification of rheumatoid arthritis and limited scleroderma as co-morbidities with microscopic colitis was noteworthy. Inflammatory arthropathy was frequently seen in microscopic colitis, usually preceding or occurring in conjunction with the onset of gastrointestinal symptoms. A variety of presentations of associated arthritis were reported: migratory, symmetric or asymmetric, peripheral or axial, oligoarticular or polyarticular, erosive or non-erosive. There was a high incidence of autoantibodies in these patients, supporting a potential autoimmune association. On the basis of these anecdotal reports, we would suggest development of a clinical registry to help define the incidence of extra-intestinal manifestations and systemic autoimmune rheumatic diseases among microscopic colitis patients to help elucidate shared predispositions, pathogenic mechanisms and therapeutic opportunities.
Collapse
Affiliation(s)
- Christopher Le
- Department of Medicine, Bayonne Medical Center, CarePoint Health, Bayonne, NJ, USA
| | - Noam Zeffren
- Department of Medicine, Bayonne Medical Center, CarePoint Health, Bayonne, NJ, USA
| | - Neil Kramer
- Institute for Rheumatic & Autoimmune Diseases, Overlook Medical Center, Atlantic Health System, Summit, USA.,Division of Rheumatology, Department of Medicine, NYU Langone Medical Center, New York, USA
| | - Elliot D Rosenstein
- Institute for Rheumatic & Autoimmune Diseases, Overlook Medical Center, Atlantic Health System, Summit, USA.,Division of Rheumatology, Department of Medicine, NYU Langone Medical Center, New York, USA
| |
Collapse
|
17
|
Predictive Significance of Two MMP-9 Promoter Polymorphisms and Acetylated c-Jun Transcription Factor for Papillary Thyroid Carcinoma Advancement. Diagnostics (Basel) 2022; 12:diagnostics12081953. [PMID: 36010303 PMCID: PMC9406990 DOI: 10.3390/diagnostics12081953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/26/2022] [Accepted: 08/06/2022] [Indexed: 11/27/2022] Open
Abstract
Papillary thyroid carcinoma represents a challenge from a prognostic standpoint. Molecular alterations responsible for PTC advancement include MMP-9 genetic promoter polymorphisms that bind transcription factors with varying degrees of affinity and, hence, constitute a predisposition for MMP-9 expression. We examined how two promoter polymorphisms (the -1562 C/T transition and -131 (CA)n tandem repeats) as well as levels of the c-Jun transcription factor and its modified form acetylated at Lys271 influence MMP-9 expression and PTC progression. A significant proportion of PTC samples were heterozygous for the (CA)n tandem repeat number, had a transcription-promoting T allele at -1562, and expressed high levels of c-Jun, acetylated c-Jun, and MMP-9 protein. The T allele at the -1562 position accompanied the elevated MMP-9 protein expression, while high acetylated c-Jun levels accompanied the high MMP-9 protein levels on mRNA. The -1562 C/T transition, MMP-9, and acetylated c-Jun were associated with the presence of extra-thyroid invasion and degree of tumor infiltration, while the T allele and acetylated c-Jun also correlated with tumor stage. We conclude that the -1562 MMP-9 polymorphism and levels of acetylated c-Jun affect PTC progression via modulation of MMP-9 levels. Genotyping the MMP-9 at -1562 and estimating the levels of MMP-9 and acetylated c-Jun in PTC may prove beneficial in identifying high-risk patients.
Collapse
|
18
|
Piperigkou Z, Koutsandreas A, Franchi M, Zolota V, Kletsas D, Passi A, Karamanos NK. ESR2 Drives Mesenchymal-to-Epithelial Transition in Triple-Negative Breast Cancer and Tumorigenesis In Vivo. Front Oncol 2022; 12:917633. [PMID: 35719919 PMCID: PMC9203970 DOI: 10.3389/fonc.2022.917633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/09/2022] [Indexed: 11/21/2022] Open
Abstract
Estrogen receptors (ERs) have pivotal roles in the development and progression of triple-negative breast cancer (TNBC). Interactions among cancer cells and tumor microenvironment are orchestrated by the extracellular matrix that is rapidly emerging as prominent contributor of fundamental processes of breast cancer progression. Early studies have correlated ERβ expression in tumor sites with a more aggressive clinical outcome, however ERβ exact role in the progression of TNBC remains to be elucidated. Herein, we introduce the functional role of ERβ suppression following isolation of monoclonal cell populations of MDA-MB-231 breast cancer cells transfected with shRNA against human ESR2 that permanently resulted in 90% reduction of ERβ mRNA and protein levels. Further, we demonstrate that clone selection results in strongly reduced levels of the aggressive functional properties of MDA-MB-231 cells, by transforming their morphological characteristics, eliminating the mesenchymal-like traits of triple-negative breast cancer cells. Monoclonal populations of shERβ MDA-MB-231 cells undergo universal matrix reorganization and pass on a mesenchymal-to-epithelial transition state. These striking changes are encompassed by the total prevention of tumorigenesis in vivo following ERβ maximum suppression and isolation of monoclonal cell populations in TNBC cells. We propose that these novel findings highlight the promising role of ERβ targeting in future pharmaceutical approaches for managing the metastatic dynamics of TNBC breast cancer.
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Anastasios Koutsandreas
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Marco Franchi
- Department for Life Quality Study, University of Bologna, Rimini, Italy
| | - Vasiliki Zolota
- Department of Pathology, School of Medicine, University of Patras, Patras, Greece
| | - Dimitrios Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biology, National Centre for Scientific Research (N.C.S.R). "Demokritos", Athens, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
19
|
Phillippi DT, Daniel S, Nguyen KN, Penaredondo BA, Lund AK. Probiotics Function as Immunomodulators in the Intestine in C57Bl/6 Male Mice Exposed to Inhaled Diesel Exhaust Particles on a High-Fat Diet. Cells 2022; 11:cells11091445. [PMID: 35563751 PMCID: PMC9101602 DOI: 10.3390/cells11091445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/10/2022] [Accepted: 04/20/2022] [Indexed: 12/04/2022] Open
Abstract
Epidemiological studies reveal a correlation between air pollution exposure and gastrointestinal (GI) diseases, yet few studies have investigated the role of inhaled particulate matter on intestinal integrity in conjunction with a high-fat (HF) diet. Additionally, there is currently limited information on probiotics in mitigating air-pollutant responses in the intestines. Thus, we investigated the hypothesis that exposure to inhaled diesel exhaust particles (DEP) and a HF diet can alter intestinal integrity and inflammation, which can be attenuated with probiotics. 4-6-w-old male C57Bl/6 mice on a HF diet (45% kcal fat) were randomly assigned to be exposed via oropharyngeal aspiration to 35 µg of DEP suspended in 35 µL of 0.9% sterile saline or sterile saline (CON) only twice a week for 4 w. A subset of mice was treated with 0.3 g/day of Winclove Ecologic® barrier probiotics (PRO) in drinking water throughout the duration of the study. Our results show that DEP exposure ± probiotics resulted in increased goblet cells and mucin (MUC)-2 expression, as determined by AB/PAS staining. Immunofluorescent quantification and/or RT-qPCR showed that DEP exposure increases claudin-3, occludin, zona occludens (ZO)-1, matrix metalloproteinase (MMP)-9, and toll-like receptor (TLR)-4, and decreases tumor necrosis factor (TNF)-α and interleukin (IL)-10 expression compared to CON. DEP exposure + probiotics increases expression of claudin-3, occludin, ZO-1, TNF-α, and IL-10 and decreases MMP-9 and TLR-4 compared to CON + PRO in the small intestine. Collectively, these results show that DEP exposure alters intestinal integrity and inflammation in conjunction with a HF diet. Probiotics proved fundamental in understanding the role of the microbiome in protecting and altering inflammatory responses in the intestines following exposure to inhaled DEP.
Collapse
Affiliation(s)
| | | | | | | | - Amie K. Lund
- Correspondence: ; Tel.: +1-(940)-369-8946; Fax: +1-(940)-565-4297
| |
Collapse
|
20
|
Groestlinger J, Spindler V, Pahlke G, Rychlik M, Del Favero G, Marko D. Alternaria alternata Mycotoxins Activate the Aryl Hydrocarbon Receptor and Nrf2-ARE Pathway to Alter the Structure and Immune Response of Colon Epithelial Cells. Chem Res Toxicol 2022; 35:731-749. [PMID: 35405071 PMCID: PMC9115800 DOI: 10.1021/acs.chemrestox.1c00364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
After ingestion of food commodities, the gastrointestinal tract (GIT) poses the first barrier against xenobiotics and pathogens. Therefore, it is regularly confronted with external stressors potentially affecting the inflammatory response and the epithelial barrier. Alternaria mycotoxins such as alternariol (AOH) and altertoxin II (ATX-II) are frequently occurring food and feed contaminants that are described for their immunomodulatory capacities. Hence, this study aimed at exploring the effect of AOH and ATX-II as single compounds or binary mixtures on the immune response and epithelial homeostasis in noncancerous colon epithelial cells HCEC-1CT. Both toxins suppressed mRNA levels of proinflammatory mediators interleukin-8 (IL-8), tumor necrosis factor α (TNF-α), and secretion of IL-8, as well as mRNA levels of the matrix metallopeptidase 2 (MMP-2). Binary combinations of AOH and ATX-II reduced the response of the single toxins. Additionally, AOH and ATX-II modified immunolocalization of transmembrane proteins such as integrin β1, zona occludens 1 (ZO-1), claudin 4 (Cldn 4), and occludin (Ocln), which support colonic tissue homeostasis and intestinal barrier function. Moreover, the cellular distribution of ZO-1 was affected by ATX-II. Mechanistically, these effects could be traced back to the involvement of several transcription factors. AOH activated the nuclear translocation of the aryl hydrocarbon receptor (AhR) and the nuclear factor erythroid 2-related factor 2 (Nrf2), governing cell metabolic competence and structural integrity. This was accompanied by altered distribution of the NF-κB p65 protein, an important regulator of inflammatory response. ATX-II also induced AhR and Nrf2 translocation, albeit failing to substantiate the effect of AOH on the colonic epithelium. Hence, both toxins coherently repress the intestinal immune response on the cytokine transcriptional and protein levels. Furthermore, both mycotoxins affected the colonic epithelial integrity by altering the cell architecture.
Collapse
Affiliation(s)
- Julia Groestlinger
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090 Vienna, Austria
| | - Veronika Spindler
- Chair of Food Analytical Chemistry, Technical University of Munich, Maximus-von-Imhof-Forum 2, 85354 Freising, Germany
| | - Gudrun Pahlke
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090 Vienna, Austria
| | - Michael Rychlik
- Chair of Food Analytical Chemistry, Technical University of Munich, Maximus-von-Imhof-Forum 2, 85354 Freising, Germany
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090 Vienna, Austria.,Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090 Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090 Vienna, Austria
| |
Collapse
|
21
|
Iacucci M, Jeffery L, Acharjee A, Nardone OM, Zardo D, Smith SCL, Bazarova A, Cannatelli R, Shivaji UN, Williams J, Gkoutos G, Ghosh S. Ultra-high Magnification Endocytoscopy and Molecular Markers for Defining Endoscopic and Histologic Remission in Ulcerative Colitis-An Exploratory Study to Define Deep Remission. Inflamm Bowel Dis 2021; 27:1719-1730. [PMID: 34019073 PMCID: PMC8528147 DOI: 10.1093/ibd/izab059] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Endoscopic and histological remission are both important treatment goals in patients with ulcerative colitis (UC). We aimed to define cellular architecture, expression of molecular markers, and their correlation with endoscopic scores assessed by ultra-high magnification endocytoscopy (ECS) and histological scores. METHODS Patients with UC (n = 29) were prospectively recruited. The correlation among ECS score (ECSS), Mayo endoscopic score (MES), and histological scores were determined. Area under curve were plotted to determine the best thresholds for ECSS that predicted histological remission by Robarts (RHI) and Nancy Histological Index (NHI).Soluble analytes relevant to inflammation were measured in serum and mucosal culture supernatants using ProcartaPlex Luminex assays and studied by partial least square discriminant analysis and logistic model. Mucosal RNA sequencing and bioinformatics analysis were performed to define differentially expressed genes/pathways. RESULTS Endocytoscope scoring system correlated strongly with RHI (r = 0.89; 95% CI, 0.51-0.98) and NHI (r = 0.86; 95% CI, 0.42-0.98) but correlated poorly with MES (r = 0.28; 95% CI, 0.27-0.70). We identified soluble brain-derived neurotrophic factors (BDNF), macrophage inflammatory proteins (MIP-1 α) and soluble vascular cell adhesion molecule 1 (sVCAM-1) predicted histological remission. Mucosal biopsy cultures also identified sVCAM-1 associated with healed mucosa. RNA-seq analysis identified gene expressions shared between ECSS, RHI, or NHI defined healing. A number of gene expressions and pathways were identified including inflammation and metabolic and tumor suppressors that discriminated healed from nonhealed mucosa. CONCLUSIONS Endocytoscopy represents an interesting tool that may sit between endoscopy and histology-but closer to the latter-identifying gene expression markers and pathways that are also identified by histology.
Collapse
Affiliation(s)
- Marietta Iacucci
- Institute of Immunology & Immunotherapy University of Birmingham, Birmingham, UK
- NIHR Wellcome Trust Clinical Research Facilities University Hospitals Birmingham NHS Trust, University of Birmingham, Birmingham, UK
- NIHR Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Louisa Jeffery
- Institute of Immunology & Immunotherapy University of Birmingham, Birmingham, UK
| | - Animesh Acharjee
- Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, UK
- Institute of Translational Medicine, University of Birmingham, Birmingham, UK
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospital Birmingham, Birmingham
| | - Olga Maria Nardone
- Institute of Immunology & Immunotherapy University of Birmingham, Birmingham, UK
| | - Davide Zardo
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Samuel C L Smith
- Institute of Immunology & Immunotherapy University of Birmingham, Birmingham, UK
| | - Alina Bazarova
- Institute for Biological Physics, University of Cologne, Cologne, Germany
| | - Rosanna Cannatelli
- Institute of Immunology & Immunotherapy University of Birmingham, Birmingham, UK
| | - Uday N Shivaji
- Institute of Immunology & Immunotherapy University of Birmingham, Birmingham, UK
- NIHR Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - John Williams
- Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, UK
- Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | - Georgios Gkoutos
- Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, UK
- Institute of Translational Medicine, University of Birmingham, Birmingham, UK
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospital Birmingham, Birmingham
- MRC Health Data Research UK (HDR), Birmingham, UK
| | - Subrata Ghosh
- Institute of Immunology & Immunotherapy University of Birmingham, Birmingham, UK
- NIHR Wellcome Trust Clinical Research Facilities University Hospitals Birmingham NHS Trust, University of Birmingham, Birmingham, UK
- NIHR Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| |
Collapse
|
22
|
Combinatorial therapy in tumor microenvironment: Where do we stand? Biochim Biophys Acta Rev Cancer 2021; 1876:188585. [PMID: 34224836 DOI: 10.1016/j.bbcan.2021.188585] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/28/2021] [Accepted: 06/23/2021] [Indexed: 01/09/2023]
Abstract
The tumor microenvironment plays a pivotal role in tumor initiation and progression by creating a dynamic interaction with cancer cells. The tumor microenvironment consists of various cellular components, including endothelial cells, fibroblasts, pericytes, adipocytes, immune cells, cancer stem cells and vasculature, which provide a sustained environment for cancer cell proliferation. Currently, targeting tumor microenvironment is increasingly being explored as a novel approach to improve cancer therapeutics, as it influences the growth and expansion of malignant cells in various ways. Despite continuous advancements in targeted therapies for cancer treatment, drug resistance, toxicity and immune escape mechanisms are the basis of treatment failure and cancer escape. Targeting tumor microenvironment efficiently with approved drugs and combination therapy is the solution to this enduring challenge that involves combining more than one treatment modality such as chemotherapy, surgery, radiotherapy, immunotherapy and nanotherapy that can effectively and synergistically target the critical pathways associated with disease pathogenesis. This review shed light on the composition of the tumor microenvironment, interaction of different components within tumor microenvironment with tumor cells and associated hallmarks, the current status of combinatorial therapies being developed, and various growing advancements. Furthermore, computational tools can also be used to monitor the significance and outcome of therapies being developed. We addressed the perceived barriers and regulatory hurdles in developing a combinatorial regimen and evaluated the present status of these therapies in the clinic. The accumulating depth of knowledge about the tumor microenvironment in cancer may facilitate further development of effective treatment modalities. This review presents the tumor microenvironment as a sweeping landscape for developing novel cancer therapies.
Collapse
|
23
|
The diverse roles of myeloid derived suppressor cells in mucosal immunity. Cell Immunol 2021; 365:104361. [PMID: 33984533 DOI: 10.1016/j.cellimm.2021.104361] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/21/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
The mucosal immune system plays a vital role in protecting the host from the external environment. Its major challenge is to balance immune responses against harmful and harmless agents and serve as a 'homeostatic gate keeper'. Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of undifferentiated cells that are characterized by an immunoregulatory and immunosuppressive phenotype. Herein we postulate that MDSCs may be involved in shaping immune responses related to mucosal immunity, due to their immunomodulatory and tissue remodeling functions. Until recently, MDSCs were investigated mainly in cancerous diseases, where they induce and contribute to an immunosuppressive and inflammatory environment that favors tumor development. However, it is now becoming clear that MDSCs participate in non-cancerous conditions such as chronic infections, autoimmune diseases, pregnancy, aging processes and immune tolerance to commensal microbiota at mucosal sites. Since MDSCs are found in the periphery only in small numbers under normal conditions, their role is highlighted during pathologies characterized by acute or chronic inflammation, when they accumulate and become activated. In this review, we describe several aspects of the current knowledge characterizing MDSCs and their involvement in the regulation of the mucosal epithelial barrier, their crosstalk with commensal microbiota and pathogenic microorganisms, and their complex interactions with a variety of surrounding regulatory and effector immune cells. Finally, we discuss the beneficial and harmful outcomes of the MDSC regulatory functions in diseases affecting mucosal tissues. We wish to illuminate the pivotal role of MDSCs in mucosal immunity, the limitations in our understanding of all the players and the intricate challenges stemming from the complex interactions of MDSCs with their environment.
Collapse
|
24
|
SEARCH FOR DIAGNOSTIC RELATIONSHIPS IN PATIENTS WITH IRRITABLE BOWEL SYNDROME AND COLORECTAL POLYPS. WORLD OF MEDICINE AND BIOLOGY 2021. [DOI: 10.26724/2079-8334-2021-1-75-74-78] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
25
|
Hämetoja H, Mäkitie A, Bäck L, Leivo I, Haglund C, Sorsa T, Hagström J. Matrix metalloproteinase-7, -8, -9, -15, and -25 in minor salivary gland adenoid cystic carcinoma. Pathol Res Pract 2020; 217:153293. [PMID: 33278774 DOI: 10.1016/j.prp.2020.153293] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 11/25/2022]
Abstract
Knowledge on the role of matrix metalloproteinases (MMPs) in adenoid cystic carcinoma (ACC) is limited. MMPs are capable of degrading almost all extracellular and pericellular components to promote invasion and metastasis. This study aimed to evaluate the immunohistochemical expression of MMP-7, -8, -9, -15, and -25 in ACC and to relate the results with clinicopathological factors and survival. The study included 68 patients with minor salivary gland ACC treated at the Helsinki University Hospital (Helsinki, Finland) in 1974-2012. Samples from 52 patients were available, consisting of 44 primary tumours and eight recurrent tumours. We scored immunostaining of MMP-7, -8, -9, -15, and -25 and analysed the immunoscore against clinical and pathological parameters using statistical correlation test. MMP-9 immunoexpression in pseudocysts of ACC and in peritumoural inflammatory cells associated with better survival and fewer treatment failures. High tumoural MMP-7 and -25 associated with better survival. High tumoural MMP-15 associated with poorer survival and high tumoural MMP-9 with advanced stage and regional recurrences. Tumour cells did not show MMP-8 immunopositivity. These results suggest that MMP-9 may contribute to ACC carcinogenesis in different roles. MMP-7, -8, and -9 can stimulate signalling pathways that may promote tissue modulation and metastatic potential. MMP-15 and -25 may reflect prognosis.
Collapse
Affiliation(s)
- Hanna Hämetoja
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Oral Pathology and Radiology, University of Turku, Turku University Hospital, Turku, Finland.
| | - Antti Mäkitie
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Division of Ear, Nose and Throat Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden; Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Leif Bäck
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ilmo Leivo
- Institute of Biomedicine, Pathology, University of Turku, Turku, Finland
| | - Caj Haglund
- Research Programs Unit, Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland; Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Timo Sorsa
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Jaana Hagström
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Oral Pathology and Radiology, University of Turku, Turku University Hospital, Turku, Finland; Research Programs Unit, Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| |
Collapse
|
26
|
Identification of Differential Intestinal Mucosa Transcriptomic Biomarkers for Ulcerative Colitis by Bioinformatics Analysis. DISEASE MARKERS 2020; 2020:8876565. [PMID: 33144895 PMCID: PMC7596466 DOI: 10.1155/2020/8876565] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/05/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023]
Abstract
Background Ulcerative colitis (UC) is a complicated disease caused by the interaction between genetic and environmental factors that affect mucosal homeostasis and triggers inappropriate immune response. The purpose of the study was to identify significant biomarkers with potential therapeutic targets and the underlying mechanisms. Methods The gene expression profiles of GSE48958, GSE73661, and GSE59071 are from the GEO database. Differentially expressed genes (DEGs) were screened by the GEO2R tool. Next, the Database for Annotation, Visualization and Integrated Discovery (DAVID) was applied to analyze gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway. Then, protein-protein interaction (PPI) was visualized by Cytoscape with Search Tool for the Retrieval of Interacting Genes (STRING). Results There were a total of 128 common DEGs genes, including 86 upregulated genes enriched in extracellular space, regulation of inflammatory response, chemokine-mediated signaling pathway, response to lipopolysaccharide, and cell proliferation, while 42 downregulated genes enriched in the integral component of the membrane, the integral component of the plasma membrane, apical plasma membrane, symporter activity, and chloride channel activity. The KEGG pathway analysis results demonstrated that DEGs were particularly enriched in cytokine-cytokine receptor interaction, TNF signaling pathway, chemokine signaling pathway, pertussis, and rheumatoid arthritis. 18 central modules of the PPI networks were selected with Cytotype MCODE. Furthermore, 18 genes were found to significantly enrich in the extracellular space, inflammatory response, chemokine-mediated signaling pathway, TNF signaling pathway, regulation of cell proliferation, and immune response via reanalysis of DAVID. Conclusion The study identified DEGs, key target genes, functional pathways, and pathway analysis of UC, which may provide potential molecular targets and diagnostic biomarkers for UC.
Collapse
|
27
|
Kunkl M, Amormino C, Frascolla S, Sambucci M, De Bardi M, Caristi S, Arcieri S, Battistini L, Tuosto L. CD28 Autonomous Signaling Orchestrates IL-22 Expression and IL-22-Regulated Epithelial Barrier Functions in Human T Lymphocytes. Front Immunol 2020; 11:590964. [PMID: 33178223 PMCID: PMC7592429 DOI: 10.3389/fimmu.2020.590964] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/22/2020] [Indexed: 12/21/2022] Open
Abstract
IL-22 is a member of the IL-10 cytokine family involved in host protection against extracellular pathogens, by promoting epithelial cell regeneration and barrier functions. Dysregulation of IL-22 production has also frequently been observed in acute respiratory distress syndrome (ARDS) and several chronic inflammatory and autoimmune diseases. We have previously described that human CD28, a crucial co-stimulatory receptor necessary for full T cell activation, is also able to act as a TCR independent signaling receptor and to induce the expression of IL-17A and inflammatory cytokines related to Th17 cells, which together with Th22 cells represent the main cellular source of IL-22. Here we characterized the role of CD28 autonomous signaling in regulating IL-22 expression in human CD4+ T cells. We show that CD28 stimulation in the absence of TCR strongly up-regulates IL-22 gene expression and secretion. As recently observed for IL-17A, we also found that CD28-mediated regulation of IL-22 transcription requires the cooperative activities of both IL-6-activated STAT3 and RelA/NF-κB transcription factors. CD28-mediated IL-22 production also promotes the barrier functions of epithelial cells by inducing mucin and metalloproteases expression. Finally, by using specific inhibitory drugs, we also identified CD28-associated class 1A phosphatidylinositol 3-kinase (PI3K) as a pivotal mediator of CD28-mediated IL-22 expression and IL-22-dependent epithelial cell barrier functions.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Carola Amormino
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Simone Frascolla
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Manolo Sambucci
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Marco De Bardi
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Silvana Caristi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Stefano Arcieri
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Luca Battistini
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| |
Collapse
|
28
|
Walter L, Canup B, Pujada A, Bui TA, Arbasi B, Laroui H, Merlin D, Garg P. Matrix metalloproteinase 9 (MMP9) limits reactive oxygen species (ROS) accumulation and DNA damage in colitis-associated cancer. Cell Death Dis 2020; 11:767. [PMID: 32943603 PMCID: PMC7498454 DOI: 10.1038/s41419-020-02959-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022]
Abstract
Colitis-associated cancer (CAC) is a subtype of colon cancer that is driven by chronic inflammation and is prevalent in chronic ulcerative colitis patients. The development of CAC is associated with the inflammation-dysplasia-carcinoma pathway which is significantly different than adenoma-carcinoma pathway of sporadic colon cancer (CRC). Matrix Metalloproteinase 9 (MMP9) is a zinc-dependent endopeptidase against extracellular matrix (ECM) proteins expressed in the gastrointestinal tract during inflammation. We have previously shown that MMP9 plays a tumor suppressor role in CAC via “MMP9-Notch1-ARF-p53 axis” pathway. The aim of this study is to determine the role of MMP9 in maintaining genomic stability in CAC. Homozygous transgenic mice with constitutive-expression of MMP9 in the colonic epithelium (TgM9) with their wild-type littermates (WT) and stably transfected HCT116 cells with/without MMP9 were used for in vivo and in vitro experiments, respectively. As ‘proof of concept’ model, nanoparticles (NPs) loaded with MMP9 siRNA were used to examine the effect of MMP9 silencing in the colonic epithelium. In CAC, colonic epithelium of TgM9 mice exhibited lower amounts of reactive oxygen species (ROS), less DNA damage, and increased expression of mismatch repair genes compared to WTs. Our study showed that MMP9 expression correlates with the reduced ROS levels, decreased DNA damage, and upregulated mismatch repair pathway. This suggests that MMP9 expression is a natural biological way to suppress CAC by limiting ROS accumulation and DNA damage in the colon. Therefore, MMP9 inhibition could be deleterious for CAC patient.
Collapse
Affiliation(s)
- Lewins Walter
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Brandon Canup
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Adani Pujada
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Tien Anh Bui
- Department of Biology, Georgia State University, Atlanta, GA, United States
| | - Behafarin Arbasi
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Hamed Laroui
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Didier Merlin
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Pallavi Garg
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States.
| |
Collapse
|
29
|
Li Y, He J, Wang F, Wang X, Yang F, Zhao C, Feng C, Li T. Role of MMP-9 in epithelial-mesenchymal transition of thyroid cancer. World J Surg Oncol 2020; 18:181. [PMID: 32698816 PMCID: PMC7376963 DOI: 10.1186/s12957-020-01958-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022] Open
Abstract
Background The purpose of this study is to explore the role and mechanism of MMP-9 in the EMT process of thyroid cancer (TC), so as to provide a basis for clinical exploration of invasion and metastasis process of TC, looking for biological markers of tumor metastasis and molecular intervention therapy. Methods Western blot and RT-PCR were employed to detect the expression of MMP-9 in human normal thyroid cell line HT-ori3 and human TC cell lines IHH-4 (PTC), FTC-133, and 8505C. Expression levels of EMT-related markers: epithelial cell marker E-cadherin and stromal cell marker Vimentin in TGF-1-induced TC cell lines were detected by Western blot and RT-PCR, respectively. The effects of MMP-9 downregulation on cell invasion and metastasis were investigated by wound-healing assay and cell invasion experiment. Results The protein and mRNA expression levels of MMP-9 in TC cell lines were increased compared with the human normal thyroid cell line HT-ori3. When TGF-β1 was added, the expression of EMT and Vimentin increased while the expression of E-cadherin decreased. Compared with the control group, the TC cells stably transfected with MMP-9 shRNA showed inhibited EMT, decreased Vimentin expression, and increased E-cadherin expression. The induction of TGF-β1 did not promote the occurrence of EMT in TC cells which were stably transformed with MMP-9 shRNA. The addition of TGF-β1 to TC cells increased the ability of the cells to migrate and invade. Compared with the control group, the migration and invasion ability of TC cells stably transfected with MMP-9 shRNA was significantly reduced, and the induction of TGF-β1 could not restore the migration and invasion ability of cells without MMP-9. Conclusions In conclusion, we found that MMP-9 can be used as a biomarker for TC, which can promote the EMT process of TGF-β1 induced TC, and thus affecting the cell migration and invasion ability.
Collapse
Affiliation(s)
- Yuanchun Li
- Department of General Surgery, The Second Affiliated Hospital of Qiqihar Medical University, No.37 Zhonghua West Road, Qiqihar, 161006, People's Republic of China.
| | - Jing He
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161006, People's Republic of China
| | - Feng Wang
- Department of General Surgery, The Second Affiliated Hospital of Qiqihar Medical University, No.37 Zhonghua West Road, Qiqihar, 161006, People's Republic of China
| | - Xin Wang
- Department of General Surgery, The Second Affiliated Hospital of Qiqihar Medical University, No.37 Zhonghua West Road, Qiqihar, 161006, People's Republic of China
| | - Fan Yang
- Clinical Pathologic Diagnosis Center, Qiqihar Medical University, Qiqihar, 161006, People's Republic of China
| | - Chunyang Zhao
- Department of General Surgery, The Second Affiliated Hospital of Qiqihar Medical University, No.37 Zhonghua West Road, Qiqihar, 161006, People's Republic of China
| | - Chunling Feng
- Department of General Surgery, The Second Affiliated Hospital of Qiqihar Medical University, No.37 Zhonghua West Road, Qiqihar, 161006, People's Republic of China
| | - Tiejun Li
- Department of Clinical Medicine, Qiqihar Medical University, Qiqihar, 161006, People's Republic of China
| |
Collapse
|
30
|
Kamar SS, Baky MH, Omar AI. The beneficial influence of rhubarb on 5-fluorouracil-induced ileal mucositis and the combined role of aquaporin-4, tumour necrosis factor-α, nuclear factor-kappa B & matrix metalloproteinase-9 in rat model: histological study. Anat Cell Biol 2020; 53:228-239. [PMID: 32647090 PMCID: PMC7343559 DOI: 10.5115/acb.20.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/28/2020] [Accepted: 03/31/2020] [Indexed: 11/27/2022] Open
Abstract
A 5-fluorouracil (5-FU) is used for cancer treatment despite its cytotoxic sequelae on healthy cells, especially the rapid proliferating ones. Intestinal mucositis is one of the most frequent chemotherapeutic debilitating sequelae. Rhubarb (Rh), an ancient herb, is known for its curing effect on gastrointestinal complications. This study aims to detect the role of aquaporin-4 (AQP-4), tumour necrosis factor-α (TNF-α), nuclear factor-kappa B (NF-κB), and matrix metalloproteinase-9 (MMP-9) in 5-FU-induced ileal histological and biochemical changes and the potential therapeutic effect of Rh water extract on these changes in rats. A 45 rats were divided into 3 groups; control, 5-FU (single intraperitoneal injection of 150 mg/kg/rat) and Rh-treated (oral 20 mg/kg/day/rat for 8 days). The change in animals’ weight, incidence of diarrhoea and AQP-4 and TNF-α values in ileal homogenates were measured. Ileal sections were subjected to hematoxylin and eosin stain, periodic acid Schiff (PAS)-reaction and MMP-9, NF-κB and AQP-4 immunohistochemical staining. A 5-FU group revealed marked ileal mucosal damage associated with a significant decrease in the mean body weight, AQP-4 level and area percent of PAS and AQP-4 positive reaction. Significant increase in the mean incidence of diarrhoea, TNF-α value and area percent of MMP-9 and NF-κB was detected. These changes were significantly corrected with Rh administration. A 5-FU resulted in severe ileal mucositis through TNF-α, NF-κB, MMP-9, and AQP-4 disturbances. Rh treatment was highly effective in preventing such mucositis.
Collapse
Affiliation(s)
- Samaa Samir Kamar
- Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mostafa Hasan Baky
- Department of Pharmacology, Faculty of Pharmacology, Egyptian Russian University, Cairo, Egypt
| | - Abeer Ibraheem Omar
- Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
31
|
Wen J, Yin P, Li L, Kang G, Ning G, Cao Y, Gao F, Su Y, Wu Y, Zhang X. Knockdown of Matrix Metallopeptidase 9 Inhibits Metastasis of Oral Squamous Cell Carcinoma Cells in a Zebrafish Xenograft Model. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4350783. [PMID: 32382550 PMCID: PMC7183015 DOI: 10.1155/2020/4350783] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/20/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023]
Abstract
Destruction of extracellular matrix (ECM) is one of the basic steps of tumor invasion and metastasis. Matrix metalloproteinase (MMP) 9, a kind of zinc-ion-dependent endopeptidase, can degrade almost all protein components in the ECM, destroy the histological barrier of tumor cell invasion, and play a key role in tumor invasion and metastasis. The role of MMP-9 in tumor invasion and metastasis has attracted increasing attention and is considered the main proteolytic enzyme in this process. Although the overexpression of MMP-9 was detected in Oral squamous cell carcinoma (OSCC) tissues, further basic studies in vivo and in vitro are needed to investigate the role of MMP-9 in OSCCs and provide scientific validation. In this research, we developed a novel OSCC zebrafish xenograft model to study the role of the MMP-9 gene in oral carcinogenesis. Firstly, the MMP-9/shRNA lentiviral clone and control virus were constructed and transfected into OSCC cells. Then, the decreasing expression of MMP-9 was verified by RT-PCR and immunocytochemistry. Cell proliferation was detected by MTT assay. Colony formation was evaluated by colony formation assay. Cell invasion was evaluated using transwell invasion assay in vitro. In addition, OSCC cells with MMP-9/shRNA knockdown and control vector were injected into zebrafish and an OSCC tumor model in zebrafish was established to evaluate invasion and metastasis in vivo. Knockdown of MMP-9 gene by shRNA could inhibit OSCC cell growth and clone formation and markedly suppress cell invasion in vitro. And the knockdown of the MMP-9 gene could also significantly decrease the metastatic distance and number of metastatic tumor cells or lesions in vivo and suppress the metastasis rate in xenografted zebrafish. Taken together, these evidences indicated that the knockdown of MMP-9 might suppress OSCC cell invasion and metastasis in vivo and in vitro. The MMP-9 gene may be a promising therapeutic target for OSCCs in the future.
Collapse
Affiliation(s)
- Jinlin Wen
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Panpan Yin
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Linwei Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Guihua Kang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Guozhu Ning
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Yu Cao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Feng Gao
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Ying Su
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Yanlin Wu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Xinyan Zhang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Howlett P, Du Bruyn E, Morrison H, Godsent IC, Wilkinson KA, Ntsekhe M, Wilkinson RJ. The immunopathogenesis of tuberculous pericarditis. Microbes Infect 2020; 22:172-181. [PMID: 32092538 DOI: 10.1016/j.micinf.2020.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/03/2020] [Indexed: 10/25/2022]
Abstract
Tuberculous pericarditis is a severe form of extrapulmonary tuberculosis and is the commonest cause of pericardial effusion in high incidence settings. Mortality ranges between 8 and 34%, and it is the leading cause of pericardial constriction in Africa and Asia. Current understanding of the disease is based on models derived from studies performed in the 1940-50s. This review summarises recent advances in the histology, microbiology and immunology of tuberculous pericarditis, with special focus on the effect of Human Immunodeficiency Virus (HIV) and the determinants of constriction.
Collapse
Affiliation(s)
- Patrick Howlett
- National Heart & Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, SW3 6LY, United Kingdom; Department of Medicine, University of Cape Town, Observatory 7925, South Africa.
| | - Elsa Du Bruyn
- Department of Medicine, University of Cape Town, Observatory 7925, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Hazel Morrison
- The Jenner Institute, University of Oxford, Old Road Campus Research Build, Roosevelt Dr, Oxford OX3 7DQ, United Kingdom
| | - Isiguzo C Godsent
- National Heart & Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, SW3 6LY, United Kingdom; Department of Medicine, Federal Teaching Hospital Abakaliki, Nigeria
| | - Katalin A Wilkinson
- Department of Medicine, University of Cape Town, Observatory 7925, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa; Francis Crick Institute, 1 Midland Rd, London NW1 1AT, United Kingdom
| | - Mpiko Ntsekhe
- Department of Medicine, University of Cape Town, Observatory 7925, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Robert J Wilkinson
- Department of Medicine, University of Cape Town, Observatory 7925, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa; Francis Crick Institute, 1 Midland Rd, London NW1 1AT, United Kingdom; Department of Infectious Diseases, Imperial College London, W2 1PG, United Kingdom
| |
Collapse
|
33
|
Salminen LE, Wilcox RR, Zhu AH, Riedel BC, Ching CRK, Rashid F, Thomopoulos SI, Saremi A, Harrison MB, Ragothaman A, Knight V, Boyle CP, Medland SE, Thompson PM, Jahanshad N. Altered Cortical Brain Structure and Increased Risk for Disease Seen Decades After Perinatal Exposure to Maternal Smoking: A Study of 9000 Adults in the UK Biobank. Cereb Cortex 2019; 29:5217-5233. [PMID: 31271414 PMCID: PMC6918926 DOI: 10.1093/cercor/bhz060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 02/20/2019] [Accepted: 02/28/2019] [Indexed: 01/08/2023] Open
Abstract
Secondhand smoke exposure is a major public health risk that is especially harmful to the developing brain, but it is unclear if early exposure affects brain structure during middle age and older adulthood. Here we analyzed brain MRI data from the UK Biobank in a population-based sample of individuals (ages 44-80) who were exposed (n = 2510) or unexposed (n = 6079) to smoking around birth. We used robust statistical models, including quantile regressions, to test the effect of perinatal smoke exposure (PSE) on cortical surface area (SA), thickness, and subcortical volumes. We hypothesized that PSE would be associated with cortical disruption in primary sensory areas compared to unexposed (PSE-) adults. After adjusting for multiple comparisons, SA was significantly lower in the pericalcarine (PCAL), inferior parietal (IPL), and regions of the temporal and frontal cortex of PSE+ adults; these abnormalities were associated with increased risk for several diseases, including circulatory and endocrine conditions. Sensitivity analyses conducted in a hold-out group of healthy participants (exposed, n = 109, unexposed, n = 315) replicated the effect of PSE on SA in the PCAL and IPL. Collectively our results show a negative, long term effect of PSE on sensory cortices that may increase risk for disease later in life.
Collapse
Affiliation(s)
- Lauren E Salminen
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| | - Rand R Wilcox
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Alyssa H Zhu
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| | - Brandalyn C Riedel
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christopher R K Ching
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| | - Faisal Rashid
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| | - Sophia I Thomopoulos
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| | - Arvin Saremi
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| | - Marc B Harrison
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| | - Anjanibhargavi Ragothaman
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| | - Victoria Knight
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| | - Christina P Boyle
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Paul M Thompson
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| | - Neda Jahanshad
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA USA
| |
Collapse
|
34
|
Wang Y, Shi L, Li J, Li L, Wang H, Yang H. Involvement of p38 MAPK pathway in benzo(a)pyrene-induced human hepatoma cell migration and invasion. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:35838-35845. [PMID: 31707611 DOI: 10.1007/s11356-019-06733-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 10/10/2019] [Indexed: 06/10/2023]
Abstract
The objective of this study was to investigate the potential role of p38 mitogen-activated protein kinases (MAPK) in benzo(a)pyrene (BaP)-induced hepatoma cell migration and invasion. Western blot assay was applied to detect the expression of proteins. qRT-PCR assay was used to measure the expression of mRNA. Wound healing assay and Transwell invasion assay were performed to evaluate cell migratory ability and cell invasive ability, respectively. Our data showed that BaP exposure increased the expression of p-p38 protein in human hepatoma HepG2 cells. Exposure to BaP facilitated HepG2 cell migration and invasion, which could be blocked by p38 MAPK inhibitors. In addition, BaP exposure induced upregulation of MMP9 mRNA expression, which was modulated by p-p38. In conclusion, p38 MAPK pathway was involved in BaP-induced hepatoma cell migration and invasion.
Collapse
Affiliation(s)
- Yadong Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, No. 105 of South Nongye Road, Zhengzhou, 450016, China.
| | - Li Shi
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Jiangmin Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, No. 105 of South Nongye Road, Zhengzhou, 450016, China
| | - Li Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, No. 105 of South Nongye Road, Zhengzhou, 450016, China
| | - Haiyu Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, No. 105 of South Nongye Road, Zhengzhou, 450016, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China.
| |
Collapse
|
35
|
Tartakover Matalon S, Ringel Y, Konikoff F, Drucker L, Pery S, Naftali T. Cannabinoid receptor 2 agonist promotes parameters implicated in mucosal healing in patients with inflammatory bowel disease. United European Gastroenterol J 2019; 8:271-283. [PMID: 32213014 DOI: 10.1177/2050640619889773] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cannabis benefits patients with inflammatory bowel disease (IBD). Cannabinoid receptors are expressed in gut immune cells and in epithelial cells of inflamed guts. Mucosal healing (MH) requires epithelial layer restoration. OBJECTIVE To analyze the effects of CB2 agonist on parameters implicated in gut inflammation and MH. METHODS Mucosal samples from areas of inflamed/uninflamed colon from 16 patients with IBD were cultured without/with cannabinoid receptor 2 (CB2) agonist (JWH-133, 10 µM, 6 hours (hr)), and analyzed for epithelial/stromal cell proliferation, apoptosis (secretome matrix metalloproteinase 9 (MMP9) activity, which impairs epithelial permeability) and interleukin-8 (IL-8) levels (n = 5-9). In addition, Caco-2 (colon carcinoma epithelial cells) were cultured with biopsy secretomes (48 hr), and analyzed for phenotype and protein markers of proliferation (proliferating cell nuclear antigen), autophagy (LC3IIB) and permeability (Zonula occludens-1) (n = 4-6). RESULTS Uninflamed tissue had higher epithelial proliferation (Ki67: 50%↑, p < 0.05), and reduced secretome MMP9 activity and IL-8 levels (>50%↓, p < 0.05) compared to inflamed tissue. Treatment with CB2 agonist had no effect on epithelial apoptosis, but increased epithelial Ki67 expression (25%), and reduced secretome MMP9 and IL-8 levels in inflamed biopsies. Secretomes of CB2-treated biopsies increased Caco-2 number, migration, proliferating cell nuclear antigen and LC3IIB expression (all, p < 0.05), but had no effect on ZO-1. CONCLUSION Using ex vivo and in vitro human models, we demonstrated that manipulating the cannabinoid system affects colon cells and secretome characteristics that facilitate MH in IBD.
Collapse
Affiliation(s)
- Shelly Tartakover Matalon
- Gastroenterology Laboratory, Meir Medical Center, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yehuda Ringel
- Gastroenterology Laboratory, Meir Medical Center, Kfar Saba, Israel.,Department of Gastroenterology and Hepatology, Meir Medical Center, Kfar Saba, Israel.,Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, 4107 Bioinformatics Building, 130 Mason Farm Road, Chapel Hill, NC, 27599-7080, USA
| | - Fred Konikoff
- Gastroenterology Laboratory, Meir Medical Center, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Gastroenterology and Hepatology, Meir Medical Center, Kfar Saba, Israel
| | - Liat Drucker
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Oncogenetics Laboratory, Meir Medical Center, Kfar Saba, Israel
| | - Shaul Pery
- Gastroenterology Laboratory, Meir Medical Center, Kfar Saba, Israel
| | - Timna Naftali
- Gastroenterology Laboratory, Meir Medical Center, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Gastroenterology and Hepatology, Meir Medical Center, Kfar Saba, Israel
| |
Collapse
|
36
|
Shi W, Zou R, Yang M, Mai L, Ren J, Wen J, Liu Z, Lai R. Analysis of Genes Involved in Ulcerative Colitis Activity and Tumorigenesis Through Systematic Mining of Gene Co-expression Networks. Front Physiol 2019; 10:662. [PMID: 31214045 PMCID: PMC6554330 DOI: 10.3389/fphys.2019.00662] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 05/09/2019] [Indexed: 12/12/2022] Open
Abstract
Ulcerative colitis (UC) is an idiopathic, chronic inflammatory disorder of the colon, characterized by continuous mucosal inflammation. Recently, some studies have considered it as part of an inflammatory bowel disease-based global network. Herein, with the aim of identifying the underlying potential genetic mechanisms involved in the development of UC, multiple algorithms for weighted correlation network analysis (WGCNA), principal component analysis (PCA), and linear models for microarray data algorithm (LIMMA) were used to identify the hub genes. The map of platelet activation, ligand-receptor interaction, calcium signaling pathway, and cAMP signaling pathway showed significant links with UC development, and the hub genes CCR7, CXCL10, CXCL9, IDO1, MMP9, and VCAM1, which are associated with immune dysregulation and tumorigenesis in biological function, were found by multiple powerful bioinformatics methods. Analysis of The Cancer Genome Atlas (TCGA) also showed that the low expression of CCR7, CXCL10, CXCL9, and MMP9 may be correlated with a poor prognosis of overall survival (OS) in colorectal cancer (CRC) patients (all p < 0.05), while no significance detected in both of IDO1 and VCAM1. In addition, low expression of CCR7, CXCL10, CXCL9, MMP9, and IDO1 may be associated with a poor prognosis in recurrence free survival (RFS) time (all p < 0.05), but no significant difference was identified in VCAM1. Moreover, the NFKB1, FLI1, and STAT1 with the highest enrichment score were detected as the master regulators of hub genes. In summary, these results indicated the central role of the hub genes of CCR7, CXCL10, CXCL9, IDO1, VCAM1, and MMP9, in response to UC progression, as well as the development of UC to CRC, thus shedding light on the molecular mechanisms involved and assisting with drug target validation.
Collapse
Affiliation(s)
- Wanting Shi
- Department of Gastroenterology, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Digestive Endoscopy Center, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Rongjun Zou
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minglei Yang
- Department of Genetics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lei Mai
- Department of Gastroenterology, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Jiangnan Ren
- Digestive Endoscopy Center, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Jialing Wen
- Guangdong Institute of Gastroenterology, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaoshi Liu
- Department of Gastroenterology, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Digestive Endoscopy Center, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Renxu Lai
- Department of Gastroenterology, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Digestive Endoscopy Center, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
37
|
Mechanisms of Compound Kushen Injection for the Treatment of Lung Cancer Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:4637839. [PMID: 31275410 PMCID: PMC6558614 DOI: 10.1155/2019/4637839] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/23/2019] [Accepted: 05/20/2019] [Indexed: 12/20/2022]
Abstract
Background Compound Kushen Injection (CKI) is a Chinese patent drug that shows good efficacy in treating lung cancer (LC). However, its underlying mechanisms need to be further clarified. Methods In this study, we adopted a network pharmacology method to gather compounds, predict targets, construct networks, and analyze biological functions and pathways. Moreover, molecular docking simulation was employed to assess the binding potential of selected target-compound pairs. Results Four networks were established, including the compound-putative target network, protein-protein interaction (PPI) network of LC targets, compound-LC target network, and herb-compound-target-pathway network. Network analysis showed that 8 targets (CHRNA3, DRD2, PRKCA, CDK1, CDK2, CHRNA5, MMP1, and MMP9) may be the therapeutic targets of CKI in LC. In addition, molecular docking simulation indicated that CHRNA3, DRD2, PRKCA, CDK1, CDK2, MMP1, and MMP9 had good binding activity with the corresponding compounds. Furthermore, enrichment analysis indicated that CKI might exert a therapeutic role in LC by regulating some important pathways, namely, pathways in cancer, proteoglycans in cancer, PI3K-Akt signaling pathway, non-small-cell lung cancer, and small cell lung cancer. Conclusions This study validated and predicted the mechanism of CKI in treating LC. Additionally, this study provides a good foundation for further experimental studies and promotes the reasonable application of CKI in the clinical treatment of LC.
Collapse
|
38
|
Monette A, Morou A, Al-Banna NA, Rousseau L, Lattouf JB, Rahmati S, Tokar T, Routy JP, Cailhier JF, Kaufmann DE, Jurisica I, Lapointe R. Failed immune responses across multiple pathologies share pan-tumor and circulating lymphocytic targets. J Clin Invest 2019; 129:2463-2479. [PMID: 30912767 DOI: 10.1172/jci125301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rationale Tumor infiltrating lymphocytes are widely associated with positive outcomes, yet carry key indicators of a systemic failed immune response against unresolved cancer. Cancer immunotherapies can reverse their tolerance phenotypes, while preserving tumor-reactivity and neoantigen-specificity shared with circulating immune cells. Objectives We performed comprehensive transcriptomic analyses to identify gene signatures common to circulating and tumor infiltrating lymphocytes in the context of clear cell renal cell carcinoma. Modulated genes also associated with disease outcome were validated in other cancer types. Findings Using bioinformatics, we identified practical diagnostic markers and actionable targets of the failed immune response. On circulating lymphocytes, three genes, LEF1, FASLG, and MMP9, could efficiently stratify patients from healthy control donors. From their associations with resistance to cancer immunotherapies and microbial infections, we uncovered not only pan-cancer, but pan-pathology failed immune response profiles. A prominent lymphocytic matrix metallopeptidase cell migration pathway, is central to a panoply of diseases and tumor immunogenicity, correlates with multi-cancer recurrence, and identifies a feasible, non-invasive approach to pan-pathology diagnoses. Conclusions The non-invasive differently expressed genes we have identified warrant future investigation towards the development of their potential in precision diagnostics and precision pan-disease immunotherapeutics.
Collapse
Affiliation(s)
- Anne Monette
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Montreal Cancer Institute, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Antigoni Morou
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Nadia A Al-Banna
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Montreal Cancer Institute, Montreal, Quebec, Canada.,Faculty of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Basic Medical Sciences, College of Medicine, QU Health Cluster, Qatar University, Doha, Qatar
| | - Louise Rousseau
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada
| | - Jean-Baptiste Lattouf
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Montreal Cancer Institute, Montreal, Quebec, Canada.,Department of Surgery, University of Montreal, Montreal, Quebec, Canada
| | - Sara Rahmati
- Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Tomas Tokar
- Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illnesses Service and Division of Hematology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Jean-François Cailhier
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Montreal Cancer Institute, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.,Nephrology Division, Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Daniel E Kaufmann
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Igor Jurisica
- Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada.,Department of Medical Biophysics and.,Department of Computer Science, University of Toronto, Toronto, Ontario, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, Slovak Republic
| | - Réjean Lapointe
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada.,Montreal Cancer Institute, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
39
|
Targeting Tumor Microenvironment for Cancer Therapy. Int J Mol Sci 2019; 20:ijms20040840. [PMID: 30781344 PMCID: PMC6413095 DOI: 10.3390/ijms20040840] [Citation(s) in RCA: 751] [Impact Index Per Article: 150.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer development is highly associated to the physiological state of the tumor microenvironment (TME). Despite the existing heterogeneity of tumors from the same or from different anatomical locations, common features can be found in the TME maturation of epithelial-derived tumors. Genetic alterations in tumor cells result in hyperplasia, uncontrolled growth, resistance to apoptosis, and metabolic shift towards anaerobic glycolysis (Warburg effect). These events create hypoxia, oxidative stress and acidosis within the TME triggering an adjustment of the extracellular matrix (ECM), a response from neighbor stromal cells (e.g., fibroblasts) and immune cells (lymphocytes and macrophages), inducing angiogenesis and, ultimately, resulting in metastasis. Exosomes secreted by TME cells are central players in all these events. The TME profile is preponderant on prognosis and impacts efficacy of anti-cancer therapies. Hence, a big effort has been made to develop new therapeutic strategies towards a more efficient targeting of TME. These efforts focus on: (i) therapeutic strategies targeting TME components, extending from conventional therapeutics, to combined therapies and nanomedicines; and (ii) the development of models that accurately resemble the TME for bench investigations, including tumor-tissue explants, “tumor on a chip” or multicellular tumor-spheroids.
Collapse
|
40
|
Abdullah ML, Hafez MM, Al-Hoshani A, Al-Shabanah O. Anti-metastatic and anti-proliferative activity of eugenol against triple negative and HER2 positive breast cancer cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:321. [PMID: 30518369 PMCID: PMC6282398 DOI: 10.1186/s12906-018-2392-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/27/2018] [Indexed: 11/13/2022]
Abstract
Background Eugenol is a natural phenolic compound and possesses anticancer and antibacterial activities. Breast cancer is a major global health problem, and most of the chemotherapeutic agents are highly toxic with long-term side effects. Therefore, this study aimed to explore the possibility of using eugenol as an anti-metastatic and anti-proliferative agent against MDA-MB-231 and SK-BR-3 breast cancer cells. Methods Breast cancer cell lines MDA-MB-231 and SK-BR-3 were treated with eugenol and cell proliferation was measured using a real-time cell electronic sensing system. Annexin V analysis with flow cytometry was used to detect the effect of eugenol on cell death. In MDA-MB-231 and SK-BR-3 cells, metastatic potential after eugenol treatment was examined using a wound-healing assay. Real-time PCR was used to study the effect of eugenol on the expression of anti-metastatic genes such as MMP2, MMP9, and TIMP-1, and genes involved in apoptosis including Caspase3, Caspase7, and Caspase9. Results Treatment with 4 μM and 8 μM eugenol for 48 h significantly inhibited cell proliferation of MDA-MB-231, with an inhibition rate of 76.4%, whereas 5 μM and 10 μM of eugenol for 48 h significantly inhibited the proliferation of SK-BR-3 cells with an inhibition rate of 68.1%. Eugenol-treated cells showed significantly decreased MMP2 and MMP9 expression and an insignificant increase in TIMP1 expression in HER2 positive and triple negative breast cancer cells. Eugenol significantly increased the proportion of MDA-MB-231 and SK-BR-3 cells in late apoptosis and increased the expression of Caspase3, Caspase7, and Caspase9. Conclusion To the best of our knowledge, this is the first study to describe the anti-metastatic effect of eugenol against MDA-MB-231 and SK-BR-3 breast cancer cell lines.
Collapse
|
41
|
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: 425] [Impact Index Per Article: 70.8] [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
|
42
|
Ronacher K, Sinha R, Cestari M. IL-22: An Underestimated Player in Natural Resistance to Tuberculosis? Front Immunol 2018; 9:2209. [PMID: 30319650 PMCID: PMC6167461 DOI: 10.3389/fimmu.2018.02209] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/06/2018] [Indexed: 12/22/2022] Open
Abstract
Approximately 10% of individuals latently infected with Mycobacterium tuberculosis (Mtb) develop active tuberculosis (TB) during their lifetime. Although it is well recognized that T-helper 1 immune responses are crucial for containing latent TB infection, the full array of host factors conferring protective immunity from TB progression are not completely understood. IL-22 is produced by cells of the innate and adaptive immune system including innate lymphoid cells, and natural killer cells as well as T lymphocytes (Th1, Th17, and Th22) and binds to its cognate receptor, the IL-22R1, which is expressed on non-hematopoietic cells such as lung epithelial cells. However, recent studies suggest that Mtb induces expression of the IL-22R1 on infected macrophages and multiple studies have indicated a protective role of IL-22 in respiratory tract infections. Reduced concentrations of circulating IL-22 in active TB compared to latent TB and decreased percentages of Mtb-specific IL-22 producing T cells in TB patients compared to controls designate this cytokine as a key player in TB immunology. More recently, it has been shown that in type 2 diabetes (T2D) and TB co-morbidity serum IL-22 concentrations are further reduced compared to TB patients without co-morbidities. However, whether a causative link between low IL-22 and increased susceptibility to TB and disease severity of TB exists remains to be established. This review summarizes the contribution of IL-22, a potentially under-appreciated key player in natural resistance to TB, at the interface between the immune response to Mtb and the lung epithelium.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Disease Resistance/immunology
- Epithelial Cells/immunology
- Epithelial Cells/microbiology
- Humans
- Interleukins/immunology
- Interleukins/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Latent Tuberculosis/blood
- Latent Tuberculosis/immunology
- Latent Tuberculosis/microbiology
- Lung/cytology
- Lung/immunology
- Lung/microbiology
- Lymphocyte Activation/immunology
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/microbiology
- Mycobacterium tuberculosis/immunology
- Receptors, Interleukin/immunology
- Receptors, Interleukin/metabolism
- Respiratory Mucosa/cytology
- Respiratory Mucosa/immunology
- Respiratory Mucosa/microbiology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- Tuberculosis, Pulmonary/blood
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/microbiology
- Interleukin-22
Collapse
Affiliation(s)
- Katharina Ronacher
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, SAMRC Centre for Tuberculosis Research, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- Infection, Immunity and Metabolism Group, Translational Research Institute, Mater Research Institute and The University of Queensland, Brisbane, QLD, Australia
| | - Roma Sinha
- Infection, Immunity and Metabolism Group, Translational Research Institute, Mater Research Institute and The University of Queensland, Brisbane, QLD, Australia
| | - Michelle Cestari
- Infection, Immunity and Metabolism Group, Translational Research Institute, Mater Research Institute and The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
43
|
Yue X, Han Z, Zhang L, Li J, Gong X. Aberrant expression of ALDH1, MMP9, Integrin αvβ3, and KiSS-1 in invasive ductal carcinoma and their clinical significance. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:3511-3522. [PMID: 31949729 PMCID: PMC6962838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/10/2018] [Indexed: 06/10/2023]
Abstract
BACKGROUND Aldehyde dehydrogenase 1 (ALDH1, a biomarker of cancer stem cells), matrix metalloproteinase 9 (MMP9, known as a matrilysin), Integrin αvβ3 (known as a biomarker of cell-matrix adhesion) and KiSS-1 (suppressor gene of tumor metastasis) are all related to cancer invasion and metastasis in many cancers. The purpose of this study was to investigate the expression of ALDH1, MMP9, Integrin αvβ3, and KiSS-1 in invasive ductal carcinoma (IDC), and their respective associations with clinical characteristics and survival in IDC. METHODS Immunohistochemical staining was used to detect the expression of ALDH1, MMP9, Integrin αvβ3, and KiSS-1 in 227 whole IDC tissue specimens. Patients' clinical and demographic data were both collected. RESULTS The expression of ALDH1, MMP9, and Integrin αvβ3 were significantly higher in IDC tissues than in the control tissues. The positive expressions of ALDH1, MMP9, and Integrin αvβ3 were positively associated with tumor grades, lymph node metastasis (LNM), tumor stages, and tumor node metastasis (TNM) stages, and inversely with overall survival (OS) and recurrence-free survival (RFS). Positive expression of KiSS-1 was negatively associated with tumor grades, LNM, tumor stages, and TNM stages, but positively with OS and RFS. A multivariate analysis demonstrated that the positive expression of ALDH1, MMP9, Integrin αvβ3, KiSS-1, ER, and HER-2, as well as TNM stages were independent prognostic factors for OS and RFS in IDC. CONCLUSIONS The expression of ALDH1, MMP9, Integrin αvβ3, and KiSS-11 should represent promising biomarkers in predicting metastasis and prognosis, as well as being potential therapeutic targets for IDC.
Collapse
Affiliation(s)
- Xicheng Yue
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical UniversityAnhui, China
| | - Zhengquan Han
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical UniversityAnhui, China
| | - Ligong Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical UniversityAnhui, China
| | - Jing Li
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical UniversityAnhui, China
| | - Xiaomeng Gong
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical UniversityAnhui, China
- Department of Pathology, Bengbu Medical UniversityAnhui, China
| |
Collapse
|