1
|
Dick JK, Sangala JA, Krishna VD, Khaimraj A, Hamel L, Erickson SM, Hicks D, Soigner Y, Covill LE, Johnson AK, Ehrhardt MJ, Ernste K, Brodin P, Koup RA, Khaitan A, Baehr C, Thielen BK, Henzler CM, Skipper C, Miller JS, Bryceson YT, Wu J, John CC, Panoskaltsis-Mortari A, Orioles A, Steiner ME, Cheeran MCJ, Pravetoni M, Hart GT. NK Cell and Monocyte Dysfunction in Multisystem Inflammatory Syndrome in Children. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1452-1466. [PMID: 39392378 PMCID: PMC11533154 DOI: 10.4049/jimmunol.2400395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a severe complication of SARS-CoV-2 infection characterized by multiorgan involvement and inflammation. Testing of cellular function ex vivo to understand the aberrant immune response in MIS-C is limited. Despite strong Ab production in MIS-C, SARS-CoV-2 nucleic acid testing can remain positive for 4-6 wk postinfection. Therefore, we hypothesized that dysfunctional cell-mediated Ab responses downstream of Ab production may be responsible for delayed clearance of viral products in MIS-C. In MIS-C, monocytes were hyperfunctional for phagocytosis and cytokine production, whereas NK cells were hypofunctional for both killing and cytokine production. The decreased NK cell cytotoxicity correlated with an NK exhaustion marker signature and systemic IL-6 levels. Potentially providing a therapeutic option, cellular engagers of CD16 and SARS-CoV-2 proteins were found to rescue NK cell function in vitro. Taken together, our results reveal dysregulation in Ab-mediated cellular responses of myeloid and NK cells that likely contribute to the immune pathology of this disease.
Collapse
Affiliation(s)
- Jenna K. Dick
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jules A. Sangala
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
| | | | - Aaron Khaimraj
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Lydia Hamel
- Division of Critical Care, Children’s Hospital and Clinics of Minnesota, Minneapolis, MN
| | - Spencer M. Erickson
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Dustin Hicks
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Yvette Soigner
- Division of Hematology, Oncology, and Transplant, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Laura E. Covill
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Alexander K. Johnson
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Michael J. Ehrhardt
- Division of Bone Marrow Transplantation and Cellular Therapy, Department of Pediatrics, M Health Fairview Masonic Children’s Hospital, Minneapolis, MN
| | - Keenan Ernste
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Petter Brodin
- Unit for Clinical Pediatrics, Department of Women’s and Children’s Health, Karolinska Institute, Solna, Sweden
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Richard A. Koup
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Alka Khaitan
- Ryan White Center for Pediatric Infectious Diseases & Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Carly Baehr
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Beth K. Thielen
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | | | - Caleb Skipper
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Jeffrey S. Miller
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Division of Hematology, Oncology, and Transplant, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Yenan T. Bryceson
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
- Division of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
- Broegelmann Laboratory, Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Jianming Wu
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN
| | - Chandy C. John
- Ryan White Center for Pediatric Infectious Diseases & Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Angela Panoskaltsis-Mortari
- Division of Bone Marrow Transplantation and Cellular Therapy, Department of Pediatrics, M Health Fairview Masonic Children’s Hospital, Minneapolis, MN
| | - Alberto Orioles
- Division of Critical Care, Children’s Hospital and Clinics of Minnesota, Minneapolis, MN
| | - Marie E. Steiner
- Divisions of Pediatric Critical Care and Pediatric Hematology/Oncology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Maxim C. J. Cheeran
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN
| | - Marco Pravetoni
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA
| | - Geoffrey T. Hart
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
2
|
Liao J, Zhang X, Xu W, Li F, Qin G. Nasal Respiratory Epithelial Adenomatoid Hamartoma: Three Case Reports and Review of the Literature. EAR, NOSE & THROAT JOURNAL 2024; 103:696-701. [PMID: 37743844 DOI: 10.1177/01455613231198996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023] Open
Abstract
Hamartomas, as non-true tumors, are commonly found in the lungs, digestive tract, and kidneys, and there are few clinical reports on nasal cases. Nasal histiocytosis accompanied by lymphatic lesions is a common feature of nasal hamartoma. Three patients with nasal cavity hamartoma were treated in our department, including 1 male and 2 females, with nasal obstruction or epistaxis. The imaging (computed tomography, magnetic resonance imaging) of 2 patients showed a soft tissue mass in the nasal cavity and no erosion of bone, while in 1 patient, the imaging was inconclusive. The excised material in all patients was found to be a soft mass. The 3 cases showed the same pattern under histological examination, and all 3 patients were diagnosed with nasal cavity respiratory epithelial adenomatoid hamartoma (REAH). Nasal hamartoma is a rare disease with nonspecific clinical manifestations and imaging features and is often misdiagnosed. Only by a better understanding of this disease can the rate of correct clinical diagnosis be improved.
Collapse
Affiliation(s)
- Jiangxue Liao
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xuge Zhang
- Department of Otolaryngology Head and Neck Surgery, Dazhou Integrated TCM & Wester Medicine Hospital, Dazhou, Sichuan, China
| | - Wei Xu
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Fei Li
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Qin
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
3
|
Seo JW, Lee YS, Jeon IS, Yu JE, Yoo JS, Koo JK, Son DJ, Yoon JS, Han SB, Yoon DY, Roh YS, Hong JT, Shim JH. IL-32γ Induced Autophagy Through Suppression of MET and mTOR Pathways in Liver Tumor Growth Inhibition. Int J Mol Sci 2024; 25:11678. [PMID: 39519229 PMCID: PMC11547131 DOI: 10.3390/ijms252111678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Interleukin-32γ (IL-32γ) has diverse functions in various malignancies. In this study, we investigated the role of IL-32γ in autophagy induction in liver cancer cells and delineated the underlying mechanisms. We found that the increased IL-32γ expression inhibited the growth, cell cycle progression, and migration of HepG2 and Hep3B cell lines; it also decreased the expression of related proteins. Furthermore, the IL-32γ overexpression induced autophagy, as indicated by the number of puncta, the expression of LC3, and the expression of autophagy-related markers. The expression levels of LAMP1, a protein essential for autophagosome formation, and colocalization with LC3 also increased. Big data analysis revealed that the expression of MET, a well-known target of autophagy, and the expression of mTOR and mTOR-related proteins were decreased by the IL-32γ overexpression. The combination treatment of MET inhibitor, cabozantinib (2 µM), and IL-32γ overexpression further increased the number of puncta, the colocalization of LC3 and LAMP1, and the expression of autophagy-related proteins. In vivo, liver tumor growth was suppressed in the IL-32γ-overexpressing mouse model, and autophagy induction was confirmed by the increased expression of LC3 and LAMP1 and the decreased expression of autophagy pathway markers (MET and mTOR). Autophagy was also decreased in the liver tumor sample of human patients. ROC curve and spearman analysis revealed that the expression levels of LC3 and IL-32γ were significantly correlated in human tumor serum and tissues. Therefore, IL-32γ overexpression induced autophagy in liver tumors through the suppression of MET and mTOR pathways critical for tumor growth inhibition.
Collapse
Affiliation(s)
- Ji-Won Seo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Yong-Sun Lee
- Ministry of Food and Drug Safety, 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju 28159, Chungbuk, Republic of Korea;
| | - In-Sook Jeon
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Ji-Eun Yu
- College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea;
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea
| | - Jun-Sang Yoo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Ja-Keun Koo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Dong-Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Jae-Suk Yoon
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea;
| | - Yoon-Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Jin-Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Jung-Hyun Shim
- College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea;
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea
| |
Collapse
|
4
|
Ritchlin CT, Rangel-Moreno J, Martino D, Isett B, Paine A, Bhattacharya S, Fox J, Meyer EM, Bao R, Bruno T, Tausk F, de la Luz Garcia-Hernandez M. Psoriatic arthritis subtypes are phenocopied in humanized mice. JCI Insight 2024; 9:e178213. [PMID: 39114979 PMCID: PMC11383598 DOI: 10.1172/jci.insight.178213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 06/26/2024] [Indexed: 09/13/2024] Open
Abstract
Psoriatic arthritis (PsA) is a complex inflammatory disease that challenges diagnosis and complicates the rational selection of effective therapies. Although T cells are considered active effectors in psoriasis and PsA, the role of CD8+ T cells in pathogenesis is not well understood. We selected the humanized mouse model NSG-SGM3 transgenic strain to examine psoriasis and PsA endotypes. Injection of PBMCs and sera from patients with psoriasis and PsA generated parallel skin and joint phenotypes in the recipient mouse. The transfer of human circulating memory T cells was followed by migration and accumulation in the skin and synovia of these immunodeficient mice. Unexpectedly, immunoglobulins were required for recapitulation of the clinical phenotype of psoriasiform lesions and PsA domains (dactylitis, enthesitis, bone erosion). Human CD8+ T cells expressing T-bet, IL-32 and CXCL14 were detected by spatial transcriptomics in murine synovia and by immunofluorescence in the human PsA synovia. Importantly, depletion of human CD8+ T cells prevented skin and synovial inflammation in mice humanized with PsA peripheral blood cells. The humanized model of psoriasis and PsA represents a valid platform for accelerating the understanding of disease pathogenesis, improving the design of personalized therapies, and revealing psoriatic disease targets.
Collapse
Affiliation(s)
| | | | - Delaney Martino
- University of Rochester Medical Center, Rochester, New York, USA
| | - Brian Isett
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Ananta Paine
- University of Rochester Medical Center, Rochester, New York, USA
| | | | - Jeffrey Fox
- Center for Musculoskeletal Research, University of Rochester Medical Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Ernest M Meyer
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Riyue Bao
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine and
| | - Tullia Bruno
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Francisco Tausk
- University of Rochester Medical Center, Rochester, New York, USA
| | | |
Collapse
|
5
|
Li N, Jia W, Wang J, Shao Q, Feng X, Li Z, Sun W, Kang M, Hu D, Xing L, Zhan X. Clinically relevant immune subtypes based on alternative splicing landscape of immune-related genes for lung cancer advanced PPPM approach. EPMA J 2024; 15:345-373. [PMID: 38841624 PMCID: PMC11147996 DOI: 10.1007/s13167-024-00366-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024]
Abstract
Background Alternative splicing (AS) occurs in the process of gene post-transcriptional process, which is very important for the correct synthesis and function of protein. The change of AS pattern may lead to the change of expression level or function of lung cancer-related genes, and then affect the occurrence and development of lung cancers. The specific AS pattern might be used as a biomarker for early warning and prognostic assessment of a cancer in the framework of predictive, preventive, and personalized medicine (PPPM; 3PM). AS events of immune-related genes (IRGs) were closely associated with tumor progression and immunotherapy. We hypothesize that IRG-AS events are significantly different in lung adenocarcinomas (LUADs) vs. controls or in lung squamous cell carcinomas (LUSCs) vs. controls. IRG-AS alteration profiling was identified to construct IRG-differentially expressed AS (IRG-DEAS) signature models. Study on the selective AS events of specific IRGs in lung cancer patients might be of great significance for further exploring the pathogenesis of lung cancer, realizing early detection and effective monitoring of lung cancer, finding new therapeutic targets, overcoming drug resistance, and developing more effective therapeutic strategies, and better used for the prediction, diagnosis, prevention, and personalized medicine of lung cancer. Methods The transcriptomic, clinical, and AS data of LUADs and LUSCs were downloaded from TCGA and its SpliceSeq databases. IRG-DEAS events were identified in LUAD and LUSC, followed by their functional characteristics, and overall survival (OS) analyses. OS-related IRG-DEAS prognostic models were constructed for LUAD and LUSC with Lasso regression, which were used to classify LUADs and LUSCs into low- and high-risk score groups. Furthermore, the immune cell distribution, immune-related scores, drug sensitivity, mutation status, and GSEA/GSVA status were analyzed between low- and high-risk score groups. Also, low- and high-immunity clusters and AS factor (SF)-OS-related-AS co-expression network and verification of cell function of CELF6 were analyzed in LUAD and LUSC. Results Comprehensive analysis of transcriptomic, clinical, and AS data of LUADs and LUSCs identified IRG-AS events in LUAD (n = 1607) and LUSC (n = 1656), including OS-related IRG-AS events in LUAD (n = 127) and LUSC (n = 105). A total of 66 IRG-DEAS events in LUAD and 89 IRG-DEAS events in LUSC were identified compared to controls. The overlapping analysis between IRG-DEASs and OS-related IRG-AS events revealed 14 OS-related IRG-DEAS events for LUAD and 16 OS-related IRG-DEAS events for LUSC, which were used to identify and optimize a 12-OS-related-IRG-DEAS signature prognostic model for LUAD and an 11-OS-related-IRG-DEAS signature prognostic model for LUSC. These two prognostic models effectively divided LUAD or LUSC samples into low- and high-risk score groups that were closely associated with OS, clinical characteristics, and tumor immune microenvironment, with significant gene sets and pathways enriched in the two groups. Moreover, weighted gene co-expression network (WGCNA) and nonnegative matrix factorization method (NMF) analyses identified four OS-relevant subtypes of LUAD and six OS-relevant subtypes of LUSC, and ssGSEA identified five immunity-relevant subtypes of LUAD and five immunity-relevant subtypes of LUSC. Interestingly, splicing factors-OS-related-AS network revealed hub molecule CELF6 was significantly related to the malignant phenotype in lung cancer cells. Conclusions This study established two reliable IRG-DEAS signature prognostic models and constructed interesting splicing factor-splicing event networks in LUAD and LUSC, which can be used to construct clinically relevant immune subtypes, patient stratification, prognostic prediction, and personalized medical services in the PPPM practice. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00366-4.
Collapse
Affiliation(s)
- Na Li
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Wenshuang Jia
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Jiahong Wang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Baiyun Road 1083, Guangzhou, Guangdong 510515 People’s Republic of China
| | - Qianwen Shao
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xiaoxia Feng
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Zhijun Li
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Wenhao Sun
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Ming Kang
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Dongming Hu
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Ligang Xing
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
6
|
Jo H, Shin S, Agura T, Jeong S, Ahn H, Lee J, Kim Y, Kang JS. The Role of α-Enolase on the Production of Interleukin (IL)-32 in Con A-Mediated Inflammation and Rheumatoid Arthritis (RA). Pharmaceuticals (Basel) 2024; 17:531. [PMID: 38675491 PMCID: PMC11054489 DOI: 10.3390/ph17040531] [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: 03/22/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Interleukin (IL)-32 is produced by T lymphocytes, natural killer cells, monocytes, and epithelial cells. IL-32 induces the production of pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α, IL-1β, IL-6, and IL-8, and IL-32 expression is highly increased in rheumatoid arthritis (RA) patients. Enolase-1 (ENO1) is a glycolytic enzyme and the stimulation of ENO1 induces high levels of pro-inflammatory cytokines in concanavalin A (Con A)-activated peripheral blood mononuclear cells (PBMCs) and macrophages in RA patients. In addition, there are many reports that anti-ENO1 antibody is correlated with the disease progression of RA. It implies that ENO1 could regulate IL-32 production during inflammation related to the pathogenesis of RA. Therefore, we investigated the role of ENO1 in IL-32 production using Con A-activated PBMCs and RA PBMCs. IL-32 expression is increased by ENO1 stimulation using real-time PCR and ELISA. In addition, we confirmed that IL-32 production was decreased in Con A-activated PBMCs and RA PBMCs pre-treated with NF-κB or p38 MAPK pathway inhibitors. Taken together, these results suggest that ENO1 plays an important role in inflammation through the induction of IL-32 production by the activation of the NF-κB and p38 MAPK pathways.
Collapse
Affiliation(s)
- Hyejung Jo
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (H.J.); (T.A.); (S.J.); (H.A.); (J.L.); (Y.K.)
| | - Seulgi Shin
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea;
| | - Tomoyo Agura
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (H.J.); (T.A.); (S.J.); (H.A.); (J.L.); (Y.K.)
| | - Seoyoun Jeong
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (H.J.); (T.A.); (S.J.); (H.A.); (J.L.); (Y.K.)
| | - Hyovin Ahn
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (H.J.); (T.A.); (S.J.); (H.A.); (J.L.); (Y.K.)
| | - Junmyung Lee
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (H.J.); (T.A.); (S.J.); (H.A.); (J.L.); (Y.K.)
| | - Yejin Kim
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (H.J.); (T.A.); (S.J.); (H.A.); (J.L.); (Y.K.)
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea;
| | - Jae Seung Kang
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (H.J.); (T.A.); (S.J.); (H.A.); (J.L.); (Y.K.)
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea;
- Artificial Intelligence Institute, Seoul National University, Seoul 08826, Republic of Korea
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
7
|
Wu SS, Hung TH, Liao PS, Tsou YA, Hung YT, Lin CD, Tai CJ, Shen TC, Shih LC. Chronic Rhinosinusitis and Premorbid Gastrointestinal Tract Diseases: A Population-Based Case-Control Study. EAR, NOSE & THROAT JOURNAL 2023:1455613231218143. [PMID: 38105251 DOI: 10.1177/01455613231218143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Objectives: The primary aim was to determine the prevalence of gastrointestinal diseases in patients with chronic rhinosinusitis (CRS), utilizing the National Health Insurance Research Database (NHIRD) in Taiwan. Several studies have supported the existence of distinct immune patterns between the Asian and Western populations in CRS patients. Through the population-based case-control study, we could compare the differences between various regions and provide further treatment strategies for subsequent studies in Asian CRS patients. The secondary aim was to assess whether different types of CRS influence the correlation with specific GI diseases. Understanding how different phenotypes or endotypes of CRS may relate to distinct GI disease patterns could provide valuable insights into the underlying mechanisms and potential shared pathways between these conditions. Methods: We use the NHIRD in Taiwan. Newly diagnosed patients with CRS were selected between January 1, 2001 and December 31, 2017 as the case group, and the controls were defined as individuals without a history of CRS. Patients with CRS were divided into two groups: with nasal polyps and without nasal polyps. We also separated GI tract diseases into four groups based on their different pathophysiologies. Results: This study included 356,245 participants (CRS: 71,249 and control: 284,996). The results showed that CRS was significantly associated with some specific GI tract diseases, including acute/chronic hepatitis B, gastroesophageal reflux disease (GERD) with/without esophagitis, achalasia of cardia, peptic/gastrojejunal ulcer, Crohn's disease, and ulcerative colitis. In addition, when CRS was subcategorized into chronic rhinosinusitis with nasal polyps (CRSwNP) and chronic rhinosinusitis without nasal polyps (CRSsNP), GERD with esophagitis and peptic ulcer were significantly associated with CRSsNP. Conclusions: A significant association between CRS and premorbid GI tract diseases has been identified. Remarkably, GERD with esophagitis and peptic ulcer were significantly associated with CRSsNP. The underlying mechanisms require further investigation and may lead to new treatments for CRS. Researchers can further investigate the mechanisms by referring to our classification method to determine the implications for diagnosis and treatment.
Collapse
Affiliation(s)
- Sen-Sen Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Tzu-Hsun Hung
- Department of Otorhinolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Pei-Shao Liao
- Department of Otorhinolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yung-An Tsou
- Department of Otorhinolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Otorhinolaryngology-Head and Neck Surgery, Asia University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Yu-Tung Hung
- School of Medicine, China Medical University, Taichung, Taiwan
- Management office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Der Lin
- Department of Otorhinolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Jaan Tai
- Department of Otorhinolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Te-Chun Shen
- School of Medicine, China Medical University, Taichung, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Liang-Chun Shih
- Department of Otorhinolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Otorhinolaryngology-Head and Neck Surgery, Asia University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
8
|
Miraki Feriz A, Bahraini F, Khosrojerdi A, Azarkar S, Sajjadi SM, HosseiniGol E, Honardoost MA, Saghafi S, Silvestris N, Leone P, Safarpour H, Racanelli V. Deciphering the immune landscape of head and neck squamous cell carcinoma: A single-cell transcriptomic analysis of regulatory T cell responses to PD-1 blockade therapy. PLoS One 2023; 18:e0295863. [PMID: 38096229 PMCID: PMC10721039 DOI: 10.1371/journal.pone.0295863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023] Open
Abstract
Immunotherapy is changing the Head and Neck Squamous Cell Carcinoma (HNSCC) landscape and improving outcomes for patients with recurrent or metastatic HNSCC. A deeper understanding of the tumor microenvironment (TME) is required in light of the limitations of patients' responses to immunotherapy. Here, we aimed to examine how Nivolumab affects infiltrating Tregs in the HNSCC TME. We used single-cell RNA sequencing data from eight tissues isolated from four HNSCC donors before and after Nivolumab treatment. Interestingly, the study found that Treg counts and suppressive activity increased following Nivolumab therapy. We also discovered that changes in the CD44-SSP1 axis, NKG2C/D-HLA-E axis, and KRAS signaling may have contributed to the increase in Treg numbers. Furthermore, our study suggests that decreasing the activity of the KRAS and Notch signaling pathways, and increasing FOXP3, CTLA-4, LAG-3, and GZMA expression, may be mechanisms that enhance the killing and suppressive capacity of Tregs. Additionally, the result of pseudo-temporal analysis of the HNSCC TME indicated that after Nivolumab therapy, the expression of certain inhibitory immune checkpoints including TIGIT, ENTPD1, and CD276 and LY9, were decreased in Tregs, while LAG-3 showed an increased expression level. The study also found that Tregs had a dense communication network with cluster two, and that certain ligand-receptor pairs, including SPP1/CD44, HLA-E/KLRC2, HLA-E/KLRK1, ANXA1/FPR3, and CXCL9/FCGR2A, had notable changes after the therapy. These changes in gene expression and cell interactions may have implications for the role of Tregs in the TME and in response to Nivolumab therapy.
Collapse
Affiliation(s)
- Adib Miraki Feriz
- Student Research Committee, Birjand University of Medical Sciences (BUMS), Birjand, Iran
| | - Fatemeh Bahraini
- Student Research Committee, Birjand University of Medical Sciences (BUMS), Birjand, Iran
| | | | - Setareh Azarkar
- Student Research Committee, Birjand University of Medical Sciences (BUMS), Birjand, Iran
| | | | - Edris HosseiniGol
- Department of Computer Engineering, University of Birjand, Birjand, Iran
| | - Mohammad Amin Honardoost
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Samira Saghafi
- Cellular and Molecular Research Center (CMRC), BUMS, Birjand, Iran
- Department of Internal Medicine, School of Medicine, BUMS, Birjand, Iran
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Patrizia Leone
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | | | - Vito Racanelli
- Centre for Medical Sciences (CISMed), University of Trento and Internal Medicine Division, Santa Chiara Hospital, Provincial Health Care Agency (APSS), Trento, Italy
| |
Collapse
|
9
|
Cantelli BA, Segura GG, Bitencourt TA, de Abreu MH, Petrucelli MF, Peronni K, Sanches PR, Beleboni RO, da Silva Junior WA, Martinez-Rossi NM, Marins M, Fachin AL. Transcriptome Analysis of Co-Cultures of THP-1 Human Macrophages with Inactivated Germinated Trichophyton rubrum Conidia. J Fungi (Basel) 2023; 9:jof9050563. [PMID: 37233274 DOI: 10.3390/jof9050563] [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: 03/14/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
Although most mycoses are superficial, the dermatophyte Trichophyton rubrum can cause systemic infections in patients with a weakened immune system, resulting in serious and deep lesions. The aim of this study was to analyze the transcriptome of a human monocyte/macrophage cell line (THP-1) co-cultured with inactivated germinated T. rubrum conidia (IGC) in order to characterize deep infection. Analysis of macrophage viability by lactate dehydrogenase quantification showed the activation of the immune system after 24 h of contact with live germinated T. rubrum conidia (LGC). After standardization of the co-culture conditions, the release of the interleukins TNF-α, IL-8, and IL-12 was quantified. The greater release of IL-12 was observed during co-culturing of THP-1 with IGC, while there was no change in the other cytokines. Next-generation sequencing of the response to T. rubrum IGC identified the modulation of 83 genes; of these, 65 were induced and 18 were repressed. The categorization of the modulated genes showed their involvement in signal transduction, cell communication, and immune response pathways. In total, 16 genes were selected for validation and Pearson's correlation coefficient was 0.98, indicating a high correlation between RNA-seq and qPCR. Modulation of the expression of all genes was similar for LGC and IGC co-culture; however, the fold-change values were higher for LGC. Due to the high expression of the IL-32 gene in RNA-seq, we quantified this interleukin and observed an increased release in co-culture with T. rubrum. In conclusion, the macrophages-T. rubrum co-culture model revealed the ability of these cells to modulate the immune response, as demonstrated by the release of proinflammatory cytokines and the RNA-seq gene expression profile. The results obtained permit to identify possible molecular targets that are modulated in macrophages and that could be explored in antifungal therapies involving the activation of the immune system.
Collapse
Affiliation(s)
- Bruna Aline Cantelli
- Biotechnology Unit, University of Ribeirão Preto-UNAERP, Ribeirao Preto 14096-900, Brazil
| | | | - Tamires Aparecida Bitencourt
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto 14049-900, Brazil
| | | | - Monise Fazolin Petrucelli
- Biotechnology Unit, University of Ribeirão Preto-UNAERP, Ribeirao Preto 14096-900, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto 14096-900, Brazil
| | - Kamila Peronni
- National Institute of Science and Technology in Stem Cell and Cell Therapy, Center for Cell-Based Therapy, Ribeirao Preto 14049-900, Brazil
| | - Pablo Rodrigo Sanches
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto 14096-900, Brazil
| | - Rene Oliveira Beleboni
- Biotechnology Unit, University of Ribeirão Preto-UNAERP, Ribeirao Preto 14096-900, Brazil
- Medicine School, University of Ribeirão Preto-UNAERP, Ribeirao Preto 14096-900, Brazil
| | | | - Nilce Maria Martinez-Rossi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto 14096-900, Brazil
| | - Mozart Marins
- Biotechnology Unit, University of Ribeirão Preto-UNAERP, Ribeirao Preto 14096-900, Brazil
- Medicine School, University of Ribeirão Preto-UNAERP, Ribeirao Preto 14096-900, Brazil
| | - Ana Lúcia Fachin
- Biotechnology Unit, University of Ribeirão Preto-UNAERP, Ribeirao Preto 14096-900, Brazil
- Medicine School, University of Ribeirão Preto-UNAERP, Ribeirao Preto 14096-900, Brazil
| |
Collapse
|
10
|
Fang X, Xu J, Jin K, Qian J. Combining of immunotherapeutic approaches with chemotherapy for treatment of gastric cancer: Achievements and limitations. Int Immunopharmacol 2023; 118:110062. [PMID: 36965367 DOI: 10.1016/j.intimp.2023.110062] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/09/2023] [Accepted: 03/19/2023] [Indexed: 03/27/2023]
Abstract
Evidence reveals that gastric cancer (GC) is the fifth most common malignancy in humans, and about 770,000 people die from this cancer yearly. It has been reported that new cases and deaths from GC are more common in men than women. Therapeutic approaches, such as surgery, chemotherapy, and radiotherapy, have been common for treating GC. Nevertheless, due to the complications and limitations of these methods, researchers use novel approaches, such as immunotherapeutic or target therapies, to evaluate the effectiveness of treatment in patients with metastatic GC. Studies have shown that monotherapy is usually associated with unpromising outcomes, and combination therapy can be a more practical option for treating metastatic GC. Therefore, to clarify different aspects of chemotherapy and immunotherapy in patients with metastatic GC, this review discussed the achievements and challenges of combining immunotherapeutic methods with chemotherapeutic agents.
Collapse
Affiliation(s)
- Xingliang Fang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Jinfang Xu
- Department of Emergency Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, China
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People's Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang, Zhejiang 312500, China.
| |
Collapse
|
11
|
Single-Cell RNA-Seq Analysis Reveals Macrophages Are Involved in the Pathogenesis of Human Sporadic Acute Type A Aortic Dissection. Biomolecules 2023; 13:biom13020399. [PMID: 36830768 PMCID: PMC9952989 DOI: 10.3390/biom13020399] [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: 09/26/2022] [Revised: 01/26/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
Macrophages play an important role in the progression of sporadic acute type A aortic dissection (ATAAD). The aim of this study was to characterize the cellular heterogeneity of macrophages in ATAAD tissues by scRNA-seq. Ascending aortic wall tissue from six ATAAD patients and three heart transplant donors was assessed by scRNA-seq and then analyzed and validated by various bioinformatic algorithms and histopathology experiments. The results revealed that the proportion of macrophages in ATAAD tissues (24.51%) was significantly higher than that in normal tissues (13.69%). Among the six macrophage subclusters, pro-inflammatory macrophages accounted for 14.96% of macrophages in the AD group and 0.18% in the normal group. Chemokine- and inflammation-related genes (CCL2, CCL20, S100A8, and S100A9) were expressed more intensively in macrophages in ATAAD tissue than in those in normal tissue. Additionally, intercellular communication analysis and transcription factor analysis indicated the activation of inflammation and degradation of the extracellular matrix in ATAAD tissue. Finally, immunohistochemistry, immunofluorescence, and Western blot experiments confirmed the overexpression of macrophage marker genes (CD68 and CD163) and matrix metalloproteinases (MMP9 and MMP2) in ATAAD tissue. Collectively, our study provides a preliminary evaluation of the role of macrophages in ATAAD, and the results could aid in the development of therapeutic options in the future.
Collapse
|
12
|
Kwon OC, Park MC, Kim YG. Interleukin-32 as a biomarker in rheumatic diseases: A narrative review. Front Immunol 2023; 14:1140373. [PMID: 36875066 PMCID: PMC9974820 DOI: 10.3389/fimmu.2023.1140373] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Interleukin-32 (IL-32) is an important cytokine involved in the innate and adaptive immune responses. The role of IL-32 has been studied in the context of various diseases. A growing body of research has investigated the role of IL-32 in rheumatic diseases including inflammatory arthritides (rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis) and connective tissue diseases (systemic lupus erythematosus, systemic sclerosis, granulomatosis and polyangiitis, and giant cell arteritis). IL-32 has been shown to play different roles according to the type of rheumatic diseases. Hence, the putative role of IL-32 as a biomarker is also different in each rheumatic disease: IL-32 could serve as a biomarker for disease activity in some diseases, whereas in other diseases it could be a biomarker for certain disease manifestations. In this narrative review, we summarize the associations between IL-32 and various rheumatic diseases and discuss the putative role of IL-32 as a biomarker in each disease.
Collapse
Affiliation(s)
- Oh Chan Kwon
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min-Chan Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan, College of Medicine, Asan Medical Center, Seoul, Republic of Korea
- Convergence Medicine Research Center, Asan Institution for Life Science, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
13
|
Dhillon I, Ham J, Maul X, Rosenfeld G, Thamboo A. A pilot prospective prevalence study of chronic rhinosinusitis associated with inflammatory bowel disease. World J Otorhinolaryngol Head Neck Surg 2022; 8:339-344. [PMID: 36474669 PMCID: PMC9714050 DOI: 10.1002/wjo2.17] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 11/05/2021] [Indexed: 09/10/2024] Open
Abstract
Background Inflammatory bowel disease (IBD) is characterized by chronic inflammation of the gastrointestinal tract. Extra-intestinal manifestations such as pulmonary diseases have been reported. Chronic rhinosinusitis (CRS), an inflammatory condition of the sinonasal mucosa, has been associated with several lung diseases. Given the relationship between lung and intestinal pathologies, and lung and sinus pathologies, we aimed to determine the prevalence of IBD among CRS patients. Methods Pilot prevalence study. Ninety-two CRS patients were screened for IBD symptoms from October 2018 to January 2020. Patient-reported disease symptoms and overall quality of life were evaluated using the Sino-Nasal Outcome Test 22 (SNOT-22), Short Inflammatory Bowel Disease Questionnaire (SIBDQ), and EuroQol 5 Dimension 5 Level (EQ-5D-5L) questionnaires. The Modified Lund-Kennedy (MLK) endoscopic and Lund-Mackay (LM) grading systems were used to confirm CRS diagnoses. Individuals who reported subjective symptoms of IBD were referred to a gastroenterologist clinic for further diagnostics. Results Twenty of the 92 (20.2%, 95% CI: 12.6%-29.8%) CRS patients reported symptoms of IBD and four individuals (4.26%, 95% CI: 1.17%-10.50%) were subsequently diagnosed with IBD. Compared to patients without IBD symptoms (n = 72), those with symptoms (n = 20) reported significantly worse SNOT-22 (P = 0.002), SIBDQ (P < 0.05), and EQ-5D-3L (P = 0.0063) scores. However, these patients did not exhibit significantly different MLK (P = 0.81) or LM (P = 0.04) scores. Conclusion The prevalence of IBD may be elevated among individuals with CRS relative to the general Canadian population. This pilot study suggests that CRS with IBD is associated with lower quality of life. Further cross-sectional studies with larger sample sizes are required.
Collapse
Affiliation(s)
- India Dhillon
- Division of Otolaryngology, St. Paul's Sinus CentreUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Jennifer Ham
- Division of Otolaryngology, St. Paul's Sinus CentreUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Ximena Maul
- Division of Otolaryngology, St. Paul's Sinus CentreUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Gregory Rosenfeld
- Division of GastroenterologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Andrew Thamboo
- Division of Otolaryngology, St. Paul's Sinus CentreUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
14
|
Jeffrey MP, MacPherson CW, Tompkins TA, Green-Johnson JM. Lacticaseibacillus rhamnosus R0011 secretome attenuates Salmonella enterica serovar Typhimurium secretome-induced intestinal epithelial cell monolayer damage and pro-inflammatory mediator production in intestinal epithelial cell and antigen-presenting cell co-cultures. Front Microbiol 2022; 13:980989. [PMID: 36246229 PMCID: PMC9554441 DOI: 10.3389/fmicb.2022.980989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Certain lactic acid bacteria (LAB) are associated with immune modulatory activities including down-regulation of pro-inflammatory gene transcription and expression. While host antigen-presenting cells (APCs) and intestinal epithelial cells (IEC) can interact directly with both pathogenic and commensal bacteria through innate immune pattern recognition receptors, recent evidence indicates indirect communication through secreted molecules is an important inter-domain communication mechanism. This communication route may be especially important in the context of IEC and APC interactions which shape host immune responses within the gut environment. We have previously shown that the Lacticaseibacillus rhamnosus R0011 secretome (LrS) dampens pro-inflammatory gene transcription and mediator production from Tumor Necrosis Factor-α and Salmonella enterica serovar Typhimurium secretome (STS)-challenged HT-29 IECs through the induction of negative regulators of innate immunity. However, many questions remain about interactions mediated through these bacterial-derived soluble components and the resulting host immune outcomes in the context of IEC and APC interactions. In the present study, we examined the ability of the LrS to down-regulate pro-inflammatory gene transcription and cytokine production from STS-challenged T84 human IEC and THP-1 human monocyte co-cultures. Cytokine and chemokine profiling revealed that apically delivered LrS induces apical secretion of macrophage inhibitory factor (MIF) and down-regulates STS-induced pro-inflammatory mediator secretion into the apical and basolateral chambers of the T84/THP-1 co-culture. Transcriptional profiling confirmed these results, as the LrS attenuated STS challenge-induced CXCL8 and NFκB1 expression in T84 IECs and THP-1 APCs. Interestingly, the LrS also reversed STS-induced damage to monolayer transepithelial resistance (TER) and permeability, results which were confirmed by ZO-1 gene expression and immunofluorescence visualization of ZO-1 expression in T84 IEC monolayers. The addition of a MIF-neutralizing antibody abrogated the ability of the LrS to reverse STS-induced damage to T84 IEC monolayer integrity, suggesting a novel role for MIF in maintaining IEC barrier function and integrity in response to soluble components derived from LAB. The results presented here provide mechanistic evidence for indirect communication mechanisms used by LAB to modulate immune responses to pathogen challenge, using in vitro approaches which allow for IEC and APC cell communication in a context which more closely mimics that which occurs in vivo.
Collapse
Affiliation(s)
- Michael P. Jeffrey
- Applied Bioscience Graduate Program and the Faculty of Science, Ontario Tech University, Oshawa, ON, Canada
| | | | | | - Julia M. Green-Johnson
- Applied Bioscience Graduate Program and the Faculty of Science, Ontario Tech University, Oshawa, ON, Canada
- *Correspondence: Julia M. Green-Johnson,
| |
Collapse
|
15
|
Li J, Mao M, Li J, Chen Z, Ji Y, Kong J, Wang Z, Zhang J, Wang Y, Liang W, Liang H, Lv L, Liu Q, Yan R, Yuan H, Chen K, Chang Y, Chen G, Xing G. Oral Administration of Omega-3 Fatty Acids Attenuates Lung Injury Caused by PM2.5 Respiratory Inhalation Simply and Feasibly In Vivo. Int J Mol Sci 2022; 23:ijms23105323. [PMID: 35628131 PMCID: PMC9140442 DOI: 10.3390/ijms23105323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 02/07/2023] Open
Abstract
For developing an effective interventional approach and treatment modality for PM2.5, the effects of omega-3 fatty acids on alleviating inflammation and attenuating lung injury induced by inhalation exposure of PM2.5 were assessed in murine models. We found that daily oral administration of the active components of omega-3 fatty acids, docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA) effectively alleviated lung parenchymal lesions, restored normal inflammatory cytokine levels and oxidative stress levels in treating mice exposed to PM2.5 (20 mg/kg) every 3 days for 5 times over a 14-day period. Especially, CT images and the pathological analysis suggested protective effects of DHA and EPA on lung injury. The key molecular mechanism is that DHA and EPA can inhibit the entry and deposition of PM2.5, and block the PM2.5-mediated cytotoxicity, oxidative stress, and inflammation.
Collapse
Affiliation(s)
- Juan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
- Correspondence: (J.L.); (G.X.)
| | - Meiru Mao
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Jiacheng Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Ziteng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Ying Ji
- Institute of Textiles and Clothing, Hong Kong Polytechnic University, Hunghom, Kowloon, Hong Kong;
| | - Jianglong Kong
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Zhijie Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Jiaxin Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Yujiao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Wei Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Haojun Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Linwen Lv
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Qiuyang Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Ruyu Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Hui Yuan
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Kui Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Yanan Chang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
| | - Guogang Chen
- College of Food Science, Shihezi University, Shihezi 832000, China;
| | - Gengmei Xing
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (M.M.); (J.L.); (Z.C.); (J.K.); (Z.W.); (J.Z.); (Y.W.); (W.L.); (H.L.); (L.L.); (Q.L.); (R.Y.); (H.Y.); (K.C.); (Y.C.)
- Correspondence: (J.L.); (G.X.)
| |
Collapse
|
16
|
Houston S, Schovanek E, Conway KME, Mustafa S, Gomez A, Ramaswamy R, Haimour A, Boulanger MJ, Reynolds LA, Cameron CE. Identification and Functional Characterization of Peptides With Antimicrobial Activity From the Syphilis Spirochete, Treponema pallidum. Front Microbiol 2022; 13:888525. [PMID: 35722306 PMCID: PMC9200625 DOI: 10.3389/fmicb.2022.888525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/08/2022] [Indexed: 12/02/2022] Open
Abstract
The etiological agent of syphilis, Treponema pallidum ssp. pallidum, is a highly invasive “stealth” pathogen that can evade the host immune response and persist within the host for decades. This obligate human pathogen is adept at establishing infection and surviving at sites within the host that have a multitude of competing microbes, sometimes including pathogens. One survival strategy employed by bacteria found at polymicrobial sites is elimination of competing microorganisms by production of antimicrobial peptides (AMPs). Antimicrobial peptides are low molecular weight proteins (miniproteins) that function directly via inhibition and killing of microbes and/or indirectly via modulation of the host immune response, which can facilitate immune evasion. In the current study, we used bioinformatics to show that approximately 7% of the T. pallidum proteome is comprised of miniproteins of 150 amino acids or less with unknown functions. To investigate the possibility that AMP production is an unrecognized defense strategy used by T. pallidum during infection, we developed a bioinformatics pipeline to analyze the complement of T. pallidum miniproteins of unknown function for the identification of potential AMPs. This analysis identified 45 T. pallidum AMP candidates; of these, Tp0451a and Tp0749 were subjected to further bioinformatic analyses to identify AMP critical core regions (AMPCCRs). Four potential AMPCCRs from the two predicted AMPs were identified and peptides corresponding to these AMPCCRs were experimentally confirmed to exhibit bacteriostatic and bactericidal activity against a panel of biologically relevant Gram-positive and Gram-negative bacteria. Immunomodulation assays performed under inflammatory conditions demonstrated that one of the AMPCCRs was also capable of differentially regulating expression of two pro-inflammatory chemokines [monocyte chemoattractant protein-1 (MCP-1) and interleukin-8 (IL-8)]. These findings demonstrate proof-of-concept for our developed AMP identification pipeline and are consistent with the novel concept that T. pallidum expresses AMPs to defend against competing microbes and modulate the host immune response.
Collapse
Affiliation(s)
- Simon Houston
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Ethan Schovanek
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Kate M. E. Conway
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Sarah Mustafa
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Alloysius Gomez
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Raghavendran Ramaswamy
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Ayman Haimour
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Martin J. Boulanger
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Lisa A. Reynolds
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Caroline E. Cameron
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, United States
- *Correspondence: Caroline E. Cameron,
| |
Collapse
|
17
|
Ramírez-Sánchez AD, Chu X, Modderman R, Kooy-Winkelaar Y, Koletzko S, Korponay-Szabó IR, Troncone R, Wijmenga C, Mearin L, Withoff S, Jonkers IH, Li Y. Single-Cell RNA Sequencing of Peripheral Blood Mononuclear Cells From Pediatric Coeliac Disease Patients Suggests Potential Pre-Seroconversion Markers. Front Immunol 2022; 13:843086. [PMID: 35371081 PMCID: PMC8964997 DOI: 10.3389/fimmu.2022.843086] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/22/2022] [Indexed: 11/30/2022] Open
Abstract
Celiac Disease (CeD) is a complex immune disorder involving villous atrophy in the small intestine that is triggered by gluten intake. Current CeD diagnosis is based on late-stage pathophysiological parameters such as detection of specific antibodies in blood and histochemical detection of villus atrophy and lymphocyte infiltration in intestinal biopsies. To date, no early onset biomarkers are available that would help prevent widespread villous atrophy and severe symptoms and co-morbidities. To search for novel CeD biomarkers, we used single-cell RNA sequencing (scRNAseq) to investigate PBMC samples from 11 children before and after seroconversion for CeD and 10 control individuals matched for age, sex and HLA-genotype. We generated scRNAseq profiles of 9559 cells and identified the expected major cellular lineages. Cell proportions remained stable across the different timepoints and health conditions, but we observed differences in gene expression profiles in specific cell types when comparing patient samples before and after disease development and comparing patients with controls. Based on the time when transcripts were differentially expressed, we could classify the deregulated genes as biomarkers for active CeD or as potential pre-diagnostic markers. Pathway analysis showed that active CeD biomarkers display a transcriptional profile associated with antigen activation in CD4+ T cells, whereas NK cells express a subset of biomarker genes even before CeD diagnosis. Intersection of biomarker genes with CeD-associated genetic risk loci pinpointed genetic factors that might play a role in CeD onset. Investigation of potential cellular interaction pathways of PBMC cell subpopulations highlighted the importance of TNF pathways in CeD. Altogether, our results pinpoint genes and pathways that are altered prior to and during CeD onset, thereby identifying novel potential biomarkers for CeD diagnosis in blood.
Collapse
Affiliation(s)
- Aarón D Ramírez-Sánchez
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Xiaojing Chu
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM) & TWINCORE, Joint Ventures Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Rutger Modderman
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Yvonne Kooy-Winkelaar
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Sibylle Koletzko
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München (LMU) Klinikum Munich, Munich, Germany.,Department of Pediatric Gastroenterology and Nutrition, School of Medicine Collegium Medicum University of Warmia and Mazury, Olsztyn, Poland
| | - Ilma R Korponay-Szabó
- Coeliac Disease Center, Heim Pál National Paediatric Institute, Budapest, Hungary.,Department of Paediatrics, Faculty of Medicine and Clinical Center, University of Debrecen, Debrecen, Hungary
| | - Riccardo Troncone
- Department of Medical Translational Sciences and European Laboratory for the Investigation of Food Induced Diseases, University Federico II, Naples, Italy
| | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Luisa Mearin
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Sebo Withoff
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Iris H Jonkers
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Yang Li
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM) & TWINCORE, Joint Ventures Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.,Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
18
|
Dickman J, Howell M, Hoopes R, Wang Y, Dickerson TJ, Bottomley M, Shamma HN, Rapp CM, Turner MJ, Rohan CA, Travers JB. Insights into Lichen Planus Pigmentosus Inversus using Minimally Invasive Dermal Patch and Whole Transcriptome Analysis. JOURNAL OF CLINICAL AND INVESTIGATIVE DERMATOLOGY 2022; 10:10.13188/2373-1044.1000077. [PMID: 36003415 PMCID: PMC9397586 DOI: 10.13188/2373-1044.1000077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Lichen Planus Pigmentosus inversus (LPPi) is a rare interface and lichenoid dermatitis (ILD) and supposed variant of lichen planus (LP) that presents as well-demarcated brown to grey macules in flexural and intertriginous areas. LPPi is deemed 'inversus' because its anatomical distribution in skin folds is opposite that seen in lichen planus pigmentosus (LPP) whose pigmented lesions arise on sun-exposed skin. Biopsy is required for the clinical diagnosis of all ILDs. Though multiple clinically-oriented studies have reported differences between LPP, LPPi, and LP, few molecular studies have been performed. In this case study, 3 patients, 2 with LPPi and one with LP, provided samples using minimally invasive whole transcriptome analysis using a dermal biomarker patch. This study confirms the involvement of interferon signaling and T-cell activation in LPPi and suggests an expression profile distinct from LP. Specific genes significantly upregulated in LPPi vs LP include an intergenic splice variant of the primary pigmentation determining receptor in humans and dysregulation of genes essential for ceramide synthesis and construction of the cornified envelope. This work expands upon our knowledge of the pathogenesis of LPPi vs LP, and supports the potential use of this technology in the diagnostic clinical setting to mitigate the need for invasive procedures.
Collapse
Affiliation(s)
- Jacob Dickman
- Department of Pharmacology & Toxicology, Wright State University, Dayton Ohio
| | - Michael Howell
- Department of Pharmacology & Toxicology, Wright State University, Dayton Ohio
| | - Robert Hoopes
- Department of Pharmacology & Toxicology, Wright State University, Dayton Ohio
| | | | | | - Michael Bottomley
- Department of Mathematics and Statistics, Wright State University, Dayton Ohio
| | - H. Nicholas Shamma
- Department of Dermatology, Wright State University, Dayton Ohio
- American Dermatopathology, LLC, Centerville, Ohio
| | - Christine M. Rapp
- Department of Pharmacology & Toxicology, Wright State University, Dayton Ohio
| | - Matthew J. Turner
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana
- Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| | - Craig A. Rohan
- Department of Pharmacology & Toxicology, Wright State University, Dayton Ohio
- Department of Dermatology, Wright State University, Dayton Ohio
| | - Jeffrey B. Travers
- Department of Pharmacology & Toxicology, Wright State University, Dayton Ohio
- Department of Dermatology, Wright State University, Dayton Ohio
- Dayton Veterans Administration Medical Center, Dayton, Ohio
| |
Collapse
|
19
|
Liu Y, Zou L, Tang H, Li J, Liu H, Jiang X, Jiang B, Dong Z, Fu W. Single-Cell Sequencing of Immune Cells in Human Aortic Dissection Tissue Provides Insights Into Immune Cell Heterogeneity. Front Cardiovasc Med 2022; 9:791875. [PMID: 35433892 PMCID: PMC9008490 DOI: 10.3389/fcvm.2022.791875] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
Background Inflammation plays an important role in the progression of sporadic aortic dissection (AD). Immune cells, especially macrophages, infiltrate the aorta and secrete inflammatory cytokines and matrix metalloproteinases to cause degradation of the extracellular matrix, thereby contributing to the pathogenesis of AD. However, the cellular heterogeneity within these immune cells has not been fully characterized. Methods We used single-cell RNA sequencing to profile the transcriptomes of all immune cells in AD tissue and normal aorta. Using magnetic-activated cell sorting gating on CD45, we obtained a higher resolution identification of the immune cell subsets in the aorta. Results We observed significant differences in the proportion of major immune cell subpopulations between AD and normal aorta tissues. Macrophages accounted for a higher percentage in the normal aorta, while the proportions of T cells, B cells and natural killer (NK) cells were all increased in AD tissues. Macrophage clusters that expanded in AD tissues originated primarily from circulating monocytes and expressed genes encoding proinflammatory cytokines and molecules involved in tissue repair. T and NK cells in AD tissues exhibited enhanced cytotoxic properties. A cluster of CD4+ T cells that had expanded in AD tissues was Th17-like and might contribute to the pathogenesis of AD. Cell–cell interaction analysis highlighted the increased communication between macrophages and T cells, which primarily regulated the costimulation of T cells. Conclusions Our study provides a comprehensive characterization of immune cells in the dissected aorta with an emphasis on the role of macrophages and T cells. The information from our study improves our understanding of immune mechanisms in AD formation and helps to identify additional useful targets for early diagnosis or therapy of AD.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Lingwei Zou
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Hanfei Tang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Jie Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hao Liu
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xiaolang Jiang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Baohong Jiang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Weiguo Fu
| | - Zhihui Dong
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Zhihui Dong
| | - Weiguo Fu
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Baohong Jiang
| |
Collapse
|
20
|
Bergantini L, d'Alessandro M, Cameli P, Otranto A, Luzzi S, Bianchi F, Bargagli E. Cytokine profiles in the detection of severe lung involvement in hospitalized patients with COVID-19: The IL-8/IL-32 axis. Cytokine 2022; 151:155804. [PMID: 35063722 PMCID: PMC8765080 DOI: 10.1016/j.cyto.2022.155804] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/13/2021] [Accepted: 01/04/2022] [Indexed: 02/04/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious respiratory disorder caused by a new coronavirus called SARS-CoV-2. The pathophysiology of severe COVID-19 is associated with a "cytokine storm". IL-32 is a key modulator in the pathogenesis of various clinical conditions and is mostly induced by IL-8. IL-32 modulates important inflammatory pathways (including TNF-α, IL-6 and IL-1b), contributing to the pathogenesis of inflammatory diseases. Il-32 was never evaluated before in COVID-19 patients stratifying as mild-moderate and severe patients. A total of 64 COVID-19 patients, 27 healthy controls were consecutively enrolled in the study. Serum concentrations of biomarkers including IL-1β, IL-10, IFN-γ, TNF-α and IL-6 were quantified by bead-based multiplex analysis and Serum concentration of IL-8 and IL-32 were determined by enzyme-linked immunosorbent assay (ELISA) kits. Interestingly, among the blood parameters, neutrophil and lymphocyte counts were significantly lower in severe COVID-19 patients than in the other, on the contrary, CRP was significantly higher in severe patients than in other groups. The cytokines that best distinguished controls from COVID-19 patients were IL-8 and IL-32, while IL-6 resulted the better variables for discriminate severe group. The best model performance for severe group was obtained by the combination of IL-32, IL-6, IFN-γ, and CRP serum concentration showing an AUC = 0.83. A cut off of 15 pg/ml of IL-6 greatly discriminate survivor from death patients. New insights related to the cytokine storm in COVID-19 patients, highlighting different severity of disease infection.
Collapse
Affiliation(s)
- Laura Bergantini
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Respiratory Diseases and Transplant Unit, Siena University, Siena, Italy.
| | - Miriana d'Alessandro
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Respiratory Diseases and Transplant Unit, Siena University, Siena, Italy
| | - Paolo Cameli
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Respiratory Diseases and Transplant Unit, Siena University, Siena, Italy
| | - Ambra Otranto
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Respiratory Diseases and Transplant Unit, Siena University, Siena, Italy
| | - Simona Luzzi
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Respiratory Diseases and Transplant Unit, Siena University, Siena, Italy
| | - Francesco Bianchi
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Respiratory Diseases and Transplant Unit, Siena University, Siena, Italy
| | - Elena Bargagli
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Respiratory Diseases and Transplant Unit, Siena University, Siena, Italy
| |
Collapse
|
21
|
Shim S, Lee S, Hisham Y, Kim S, Nguyen TT, Taitt AS, Hwang J, Jhun H, Park HY, Lee Y, Yeom SC, Kim SY, Kim YG, Kim S. Comparison of the Seven Interleukin-32 Isoforms’ Biological Activities: IL-32θ Possesses the Most Dominant Biological Activity. Front Immunol 2022; 13:837588. [PMID: 35281066 PMCID: PMC8914309 DOI: 10.3389/fimmu.2022.837588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Cytokines are significantly associated with the homeostasis of immune responses in health and disease. Interleukin-32 (IL-32) is a cytokine originally discovered in natural killer cell transcript 4. IL-32 with different disorders has been described in terms of pathogenesis and the progression of diseases. Clinical studies have investigated IL-32 under various conditions, such as viral infection, autoimmune diseases, inflammatory diseases, certain types of cancer, vascular disease, and pulmonary diseases. The high expression of IL-32 was identified in different tissues with various diseases and found to have multiple transcripts of up to seven isoforms. However, the purification and biological activities of these isoforms have not been investigated yet. Therefore, in this study, we purified and compared the biological activity of recombinant IL-32 (rIL-32) isoforms. This is the first time for seven rIL-32 isoforms (α, β, δ, γ, ϵ, ζ, and θ) to be cloned and purified using an Escherichia coli expression system. Next, we evaluate the biological activities of these seven rIL-32 isoforms, which were used to treat different types of cells by assessing the levels of inflammatory cytokine production. The results revealed that rIL-32θ possessed the most dominant biological activity in both immune and non-immune cells.
Collapse
Affiliation(s)
- Saerok Shim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Siyoung Lee
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Yasmin Hisham
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Sinae Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
- College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Tam T. Nguyen
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
- College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Afeisha S. Taitt
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Jihyeong Hwang
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Hyunjhung Jhun
- Technical Assistance Center, Korea Food Research Institute, Wanju, South Korea
| | - Ho-Young Park
- Research Group of Functional Food Materials, Korea Food Research Institute, Wanju, South Korea
| | - Youngmin Lee
- Department of Medicine, Pusan Paik Hospital, Collage of Medicine, Inje University, Busan, South Korea
| | - Su Cheong Yeom
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, South Korea
| | - Sang-Yeob Kim
- Convergence Medicine Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul, South Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- *Correspondence: Yong-Gil Kim, ; Soohyun Kim,
| | - Soohyun Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
- College of Veterinary Medicine, Konkuk University, Seoul, South Korea
- *Correspondence: Yong-Gil Kim, ; Soohyun Kim,
| |
Collapse
|
22
|
Chua ZMX, Fletcher AL. The LIGHT switch: mechanisms of fibroblast pathology in eosinophilic esophagitis. Mucosal Immunol 2022; 15:195-197. [PMID: 35197559 PMCID: PMC8866120 DOI: 10.1038/s41385-022-00486-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 02/04/2023]
Affiliation(s)
- Zoe M. X. Chua
- grid.1002.30000 0004 1936 7857Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800 Australia
| | - Anne L. Fletcher
- grid.1002.30000 0004 1936 7857Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800 Australia ,grid.6572.60000 0004 1936 7486Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, B16 2TT UK
| |
Collapse
|
23
|
Gabriel EM, Wiche Salinas TR, Gosselin A, Larouche-Anctil E, Durand M, Landay AL, El-Far M, Tremblay CL, Routy JP, Ancuta P. Overt IL-32 isoform expression at intestinal level during HIV-1 infection is negatively regulated by IL-17A. AIDS 2021; 35:1881-1894. [PMID: 34101628 PMCID: PMC8416712 DOI: 10.1097/qad.0000000000002972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Untreated HIV infection was previously associated with IL-32 overexpression in gut/intestinal epithelial cells (IEC). Here, we explored IL-32 isoform expression in the colon of people with HIV (PWH) receiving antiretroviral therapy (ART) and IL-32 triggers/modulators in IEC. DESIGN Sigmoid colon biopsies (SCB) and blood were collected from ART-treated PWH (HIV + ART; n = 17; mean age: 56 years; CD4+ cell counts: 679 cells/μl; time on ART: 72 months) and age-matched HIV-uninfected controls (HIVneg; n = 5). The IEC line HT-29 was used for mechanistic studies. METHODS Cells from SCB and blood were isolated by enzymatic digestion and/or gradient centrifugation. HT-29 cells were exposed to TLR1-9 agonists, TNF-α, IL-17A and HIV. IL-32α/β/γ/D/ε/θ and IL-17A mRNA levels were quantified by real-time RT-PCR. IL-32 protein levels were quantified by ELISA. RESULTS IL-32β/γ/ε isoform transcripts were detectable in the blood and SCB, with IL-32β mRNA levels being predominantly expressed in both compartments and at significantly higher levels in HIV + ART compared to HIVneg. IL-17A transcripts were only detectable in SCB, with increased IL-17A levels in HIVneg compared with HIV + ART and negatively correlated with IL-32β mRNA levels. IL-32β/γ/ε isoform mRNA were detected in HT-29 cells upon exposure to TNF-α, Poly I:C (TLR3 agonist), Flagellin (TLR-5 agonist) and HIV. IL-17A significantly decreased both IL-32 β/γ/ε mRNA and cell-associated IL-32 protein levels induced upon TNF-α and Poly I:C triggering. CONCLUSION We document IL-32 isoforms abundant in the colon of ART-treated PWH and reveal the capacity of the Th17 hallmark cytokine IL-17A to attenuate IL-32 overexpression in a model of inflamed IEC.
Collapse
Affiliation(s)
- Etiene Moreira Gabriel
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Tomas Raul Wiche Salinas
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | | | | | - Madeleine Durand
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | | | | | - Cécile L. Tremblay
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Pierre Routy
- McGill University Health Centre, Montreal, Québec, Canada
- Chronic Viral Illness Service and Hematology Department, McGill University Health Centre, Montréal, Québec, Canada
| | - Petronela Ancuta
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
24
|
Devalraju KP, Neela VSK, Krovvidi SS, Vankayalapati R, Valluri VL. Defective expansion and function of memory like natural killer cells in HIV+ individuals with latent tuberculosis infection. PLoS One 2021; 16:e0257185. [PMID: 34516566 PMCID: PMC8437280 DOI: 10.1371/journal.pone.0257185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 08/25/2021] [Indexed: 11/29/2022] Open
Abstract
Purpose Tuberculosis (TB) is the leading cause of infectious disease related mortality, and only 10% of the infected individuals develop active disease. The likelihood of progression of latent tuberculosis infection (LTBI) to active TB disease is high in HIV infected individuals. Identification of HIV+ individuals at risk would allow treating targeted population, facilitating completion of therapy for LTBI and prevention of TB development. NK cells have an important role in T cell independent immunity against TB, but the exact role of NK cell subsets in LTBI and HIV is not well characterized. Methods In this study, we compared the expansion and function of memory like NK cells from HIV-LTBI+ individuals and treatment naïve HIV+LTBI+ patients in response to Mtb antigens ESAT-6 and CFP-10. Results In freshly isolated PBMCs, percentages of CD3-CD56+ NK cells were similar in HIV+LTBI+ patients and healthy HIV-LTBI+ individuals. However, percentages of CD3-CD56+CD16+ NK cells were higher in healthy HIV-LTBI+ individuals compared to HIV+LTBI+ patients. HIV infection also inhibited the expansion of memory like NK cells, production of IL-32α, IL-15 and IFN-γ in response to Mtb antigens in LTBI+ individuals. Conclusion We studied phenotypic, functional subsets and activation of memory like-NK cells during HIV infection and LTBI. We observed that HIV+LTBI+ patients demonstrated suboptimal NK cell and monocyte interactions in response to Mtb, leading to reduced IL-15, IFN-γ and granzyme B and increased CCL5 production. Our study highlights the effect of HIV and LTBI on modulation of NK cell activity to understand their role in development of interventions to prevent progression to TB in high risk individuals.
Collapse
Affiliation(s)
- Kamakshi Prudhula Devalraju
- Immunology and Molecular Biology Division, Blue Peter Public Health and Research Centre, LEPRA Society, Cherlapally, Hyderabad, Telangana, India
| | - Venkata Sanjeev Kumar Neela
- Immunology and Molecular Biology Division, Blue Peter Public Health and Research Centre, LEPRA Society, Cherlapally, Hyderabad, Telangana, India
| | - Siva Sai Krovvidi
- Department of Biotechnology, Sreenidhi Institute of Science and Technology, Yamnampet, Ghatkesar, Hyderabad, Telangana, India
| | - Ramakrishna Vankayalapati
- Department of Pulmonary Immunology, Center for Biomedical Research, The University of Texas Health Center at Tyler, Texas, TX, United States of America
- * E-mail: (VLV); (RV)
| | - Vijaya Lakshmi Valluri
- Immunology and Molecular Biology Division, Blue Peter Public Health and Research Centre, LEPRA Society, Cherlapally, Hyderabad, Telangana, India
- * E-mail: (VLV); (RV)
| |
Collapse
|
25
|
Fardoos R, Asowata OE, Herbert N, Nyquist SK, Zungu Y, Singh A, Ngoepe A, Mbano IM, Mthabela N, Ramjit D, Karim F, Kuhn W, Madela FG, Manzini VT, Anderson F, Berger B, Pers TH, Shalek AK, Leslie A, Kløverpris HN. HIV infection drives interferon signaling within intestinal SARS-CoV-2 target cells. JCI Insight 2021; 6:e148920. [PMID: 34252054 PMCID: PMC8409978 DOI: 10.1172/jci.insight.148920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
SARS-CoV-2 infects epithelial cells of the human gastrointestinal (GI) tract and causes related symptoms. HIV infection impairs gut homeostasis and is associated with an increased risk of COVID-19 fatality. To investigate the potential link between these observations, we analyzed single-cell transcriptional profiles and SARS-CoV-2 entry receptor expression across lymphoid and mucosal human tissue from chronically HIV-infected individuals and uninfected controls. Absorptive gut enterocytes displayed the highest coexpression of SARS-CoV-2 receptors ACE2, TMPRSS2, and TMPRSS4, of which ACE2 expression was associated with canonical interferon response and antiviral genes. Chronic treated HIV infection was associated with a clear antiviral response in gut enterocytes and, unexpectedly, with a substantial reduction of ACE2 and TMPRSS2 target cells. Gut tissue from SARS-CoV-2–infected individuals, however, showed abundant SARS-CoV-2 nucleocapsid protein in both the large and small intestine, including an HIV-coinfected individual. Thus, upregulation of antiviral response genes and downregulation of ACE2 and TMPRSS2 in the GI tract of HIV-infected individuals does not prevent SARS-CoV-2 infection in this compartment. The impact of these HIV-associated intestinal mucosal changes on SARS-CoV-2 infection dynamics, disease severity, and vaccine responses remains unclear and requires further investigation.
Collapse
Affiliation(s)
- Rabiah Fardoos
- Africa Health Research Institute, Durban, South Africa.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Osaretin E Asowata
- Africa Health Research Institute, Durban, South Africa.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Nicholas Herbert
- Africa Health Research Institute, Durban, South Africa.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sarah K Nyquist
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA.,Program in Computational and Systems Biology, MIT, Cambridge, Massachusetts, USA
| | - Yenzekile Zungu
- Africa Health Research Institute, Durban, South Africa.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Alveera Singh
- Africa Health Research Institute, Durban, South Africa
| | | | - Ian M Mbano
- Africa Health Research Institute, Durban, South Africa.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | | | | | - Farina Karim
- Africa Health Research Institute, Durban, South Africa
| | - Warren Kuhn
- ENT Department, General Justice Gizenga Mpanza Regional Hospital (Stanger Hospital), University of KwaZulu-Natal, Durban, South Africa
| | - Fusi G Madela
- Discipline of General Surgery, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban, South Africa
| | - Vukani T Manzini
- Discipline of General Surgery, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban, South Africa
| | - Frank Anderson
- Discipline of General Surgery, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban, South Africa
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory and Department of Mathematics, MIT, Cambridge, Massachusetts, USA
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alex K Shalek
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Alasdair Leslie
- Africa Health Research Institute, Durban, South Africa.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa.,Division of Infection and Immunity, University College London, London, United Kingdom
| | - Henrik N Kløverpris
- Africa Health Research Institute, Durban, South Africa.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa.,Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
26
|
Association of lncRNA PRNCR1 polymorphisms with cancer susceptibility: a meta-analysis of the current literature. J Genet 2021. [DOI: 10.1007/s12041-021-01269-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
27
|
Wu K, Zeng J, Shi X, Xie J, Li Y, Zheng H, Peng G, Zhu G, Tang D, Wu S. Targeting TIGIT Inhibits Bladder Cancer Metastasis Through Suppressing IL-32. Front Pharmacol 2021; 12:801493. [PMID: 35069212 PMCID: PMC8766971 DOI: 10.3389/fphar.2021.801493] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/24/2021] [Indexed: 02/05/2023] Open
Abstract
Bladder cancer is a highly metastatic tumor and one of the most common malignancies originating in the urinary tract. Despite the efficacy of immune checkpoints, including programmed cell death-1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), the effect of immunotherapy for bladder cancer remains unsatisfactory. Therefore, it is urgent to develop new targets to expand immunotherapeutic options. In this study, we utilized single-cell sequencing to explore the cell composition of tumors and detected a subset of Treg cells with high expression of T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) and interleukin (IL)-32. The antitumor immune response was suppressed by this subset of Treg cells, while IL-32 promoted bladder cancer metastasis. Nevertheless, targeting TIGIT not only reversed immunosuppression by restoring the antitumor immune response mediated by T cells but also suppressed the secretion of IL-32 and inhibited the metastasis of bladder cancer cells. Thus, our study provided novel insights into immunosuppression in bladder cancer and highlighted TIGIT as a novel target for immunotherapy of bladder cancer. We also illustrated the mechanism of the dual effect of targeting TIGIT and revealed the metastasis-promoting effect of IL-32 in bladder cancer. Collectively, these findings raise the possibility of utilizing TIGIT as a target against bladder cancer from the bench to the bedside.
Collapse
Affiliation(s)
- Kang Wu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Jun Zeng
- Department of Genetics and Cell Biology, College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Xulian Shi
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Jiajia Xie
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Yuqing Li
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Haoxiang Zheng
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Guoyu Peng
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Guanghui Zhu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Dongdong Tang
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Song Wu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Medical Laboratory of Shenzhen Luohu People’s Hospital, Shenzhen, China
- Teaching Center of Shenzhen Luohu Hospital, Shantou University Medical College, Shantou, China
- Department of Urology and Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Song Wu,
| |
Collapse
|
28
|
Luo F, Guo H, Yu H, Li Y, Feng Y, Wang Y. PM2.5 organic extract mediates inflammation through the ERβ pathway to contribute to lung carcinogenesis in vitro and vivo. CHEMOSPHERE 2021; 263:127867. [PMID: 32841872 DOI: 10.1016/j.chemosphere.2020.127867] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/23/2020] [Accepted: 07/26/2020] [Indexed: 06/11/2023]
Abstract
An increasing number of researches have shown that fine particulate matter (PM2.5) is closely related to increased respiratory inflammation and can even lead to lung cancer. Estrogen receptor β (ERβ) has been demonstrated to be involved in several cancers. However, the exact role of ERβ in PM2.5 organic extract (Po)-promoted inflammation and lung cancer remains unknown. The purpose of this study was to investigate whether ERβ is involved in Po induced inflammation and lung cancer. In vitro, our results showed that interleukin-6 (IL-6) and ERβ were simultaneously increased in lung bronchial epithelial cells exposed to Po; additionally, inhibition of ERβ decreased IL-6 expression and secretion through inactivating ERK and AKT and further promoted cells malignant transformation. Moreover, we performed an animal model of inhalation exposure to Po using female C57BL/6 mice. Although we were unable to find tumor tissue in mice exposed to Po, we detected evidence of lung inflammation, epithelial-to-mesenchymal transition (EMT) phenotype and severe pulmonary injury; in addition, intraperitoneal injection of PHTPP (an ERβ inhibitor) showed that the above phenomena have been improved, which demonstrate that Po stimulates IL-6 expression to promote inflammation, EMT phenotype and lung injury through the ERβ pathway. In conclusion, our results confirmed the potential toxic effect of PM2.5, and increased our understanding of PM2.5 carcinogenic potential by exploring the mechanism of ERβ regulating inflammation.
Collapse
Affiliation(s)
- Fei Luo
- Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Huaqi Guo
- Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Hengyi Yu
- Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Yan Li
- Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Yan Feng
- Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Yan Wang
- Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China; The Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.
| |
Collapse
|
29
|
Sexton RE, Al Hallak MN, Diab M, Azmi AS. Gastric cancer: a comprehensive review of current and future treatment strategies. Cancer Metastasis Rev 2020; 39:1179-1203. [PMID: 32894370 PMCID: PMC7680370 DOI: 10.1007/s10555-020-09925-3] [Citation(s) in RCA: 358] [Impact Index Per Article: 89.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
Gastric cancer remains a major unmet clinical problem with over 1 million new cases worldwide. It is the fourth most commonly occurring cancer in men and the seventh most commonly occurring cancer in women. A major fraction of gastric cancer has been linked to variety of pathogenic infections including but not limited to Helicobacter pylori (H. pylori) or Epstein Barr virus (EBV). Strategies are being pursued to prevent gastric cancer development such as H. pylori eradication, which has helped to prevent significant proportion of gastric cancer. Today, treatments have helped to manage this disease and the 5-year survival for stage IA and IB tumors treated with surgery are between 60 and 80%. However, patients with stage III tumors undergoing surgery have a dismal 5-year survival rate between 18 and 50% depending on the dataset. These figures indicate the need for more effective molecularly driven treatment strategies. This review discusses the molecular profile of gastric tumors, the success, and challenges with available therapeutic targets along with newer biomarkers and emerging targets.
Collapse
Affiliation(s)
- Rachel E Sexton
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, HWCRC 732, Detroit, MI, 48201, USA
| | - Mohammed Najeeb Al Hallak
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, HWCRC 732, Detroit, MI, 48201, USA
| | - Maria Diab
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, HWCRC 732, Detroit, MI, 48201, USA
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, HWCRC 732, Detroit, MI, 48201, USA.
| |
Collapse
|
30
|
Immunotherapeutic Potential of Interleukin-32 and Trained Immunity for Leishmaniasis Treatment. Trends Parasitol 2020; 37:130-141. [PMID: 33082090 DOI: 10.1016/j.pt.2020.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 01/23/2023]
Abstract
Neglected tropical diseases annually account for several million infections worldwide. Efficacious treatment for these poorly understood infectious diseases is often limited to ineffective, expensive, and toxic therapies such as the SbV used for leishmaniasis patients. Here, we review the latest discoveries and literature on the molecular pathways, cell types, and immune mediators involved in the immune response to infection with New World Leishmania spp. in humans and their interaction with the adaptive and innate immune system. Novel developments in the field of trained innate immunity and the recently described role of IL-32 are emphasized as potential immunotherapeutic treatments for the management of leishmaniasis.
Collapse
|
31
|
Diakowska D, Krzystek-Korpacka M. Local and Systemic Interleukin-32 in Esophageal, Gastric, and Colorectal Cancers: Clinical and Diagnostic Significance. Diagnostics (Basel) 2020; 10:E785. [PMID: 33020452 PMCID: PMC7600995 DOI: 10.3390/diagnostics10100785] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023] Open
Abstract
Little is known on clinical and diagnostic relevance of interleukin-32 in gastrointestinal tract (GIT) cancers. We determined its mRNA (n = 52) and protein (n = 63) expression in paired (tumor-normal) samples from esophageal squamous cell carcinoma (ESCC) and gastric (GC) and colorectal cancer (CRC) patients, with reference to cancer-associated genes, and quantified circulating interleukin-32 in 70 cancer patients and 28 controls. IL32 expression was significantly upregulated solely in ESCC, reflecting T stage in non-transformed tumor-adjacent tissue. Fold-change in IL32 and IL-32 was higher in left-sided CRC, owing to high interleukin expression in non-transformed right-sided colonic mucosa. IL32 was independently and positively associated with Ki67, HIF1A, and ACTA2 and negatively with TJP1 in tumors and with IL10Ra and BCLxL in non-transformed tumor-adjacent tissue. IL-32 protein was significantly upregulated in colorectal tumors. In ESCC, advanced stage and lymph node metastasis were associated with significant IL-32 upregulation. Circulating interleukin was significantly elevated in cancer patients, more so in ESCC and GC than CRC. As biomarker, IL-32 detected gastroesophageal cancers with 99.5% accuracy. In conclusion, IL-32 is upregulated in GIT cancers at local and systemic level, reflecting hypoxia and proliferative and invasive/metastatic capacity in tumors and immunosuppressive and antiapoptotic potential in non-transformed mucosa, while being an accurate biomarker of gastroesophageal cancers.
Collapse
Affiliation(s)
- Dorota Diakowska
- Department of Gastrointestinal and General Surgery, Wroclaw Medical University, 50-368 Wroclaw, Poland;
- Department of Nervous System Diseases, Wroclaw Medical University, 51-618 Wroclaw, Poland
| | | |
Collapse
|
32
|
Öngöz Dede F, Gökmenoğlu C, Deveci ET, Çelen S, Avci B, Kara C, Çanakçi V. Increased levels of interleukin-32 isoforms alpha, beta, gamma, and delta in the gingival crevicular fluid and plasma of the patients with periodontitis. J Periodontal Res 2020; 56:83-92. [PMID: 32890410 DOI: 10.1111/jre.12796] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/10/2020] [Accepted: 08/07/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND OBJECTIVE Interleukin (IL)-32, which has been recently reported to be associated with periodontitis, has been suggested to have pleiotropic effect due to its 9 different isoforms. The aim of this study was to investigate the levels of IL-32α, IL-32β, IL-32γ, IL-32δ isoforms in gingival crevicular fluid (GCF) and plasma before and after non-surgical periodontal treatment in patients with periodontitis (P). MATERIALS AND METHODS Twenty-seven P and 27 periodontally healthy controls (C) were recruited in this study. Non-surgical periodontal treatment was performed to periodontitis patients. GCF and plasma sampling and clinical periodontal parameters were evaluated before and 1 month after treatment. Enzyme-linked immunosorbent assay was used to analyze the levels of IL-32α, IL-32β, IL-32γ, IL-32δ isoforms in GCF and plasma samples. RESULTS The levels of IL-32α, IL-32β, IL-32γ, and IL-32δ in plasma and GCF were significantly higher in patients with periodontitis than healthy controls (P < .001). In P group, plasma and GCF IL-32α, IL-32β, IL-32γ, and IL-32δ levels after non-surgical periodontal treatment were lower when compared to baseline (P < .001). Moreover, there was a positive correlation between GCF and plasma IL-32α, IL-32β, IL-32γ, and IL-32δ levels in all groups at baseline and after treatment (P < .05). CONCLUSION The study supported that there was a relationship between elevated levels of IL-32 isoforms and periodontitis. Also, our novel findings suggest that the pro-inflammatory role of IL-32 in the periodontitis may be originated from IL-32α, IL-32β, IL-32γ, and IL-32δ isoforms.
Collapse
Affiliation(s)
- Figen Öngöz Dede
- Department of Periodontology, Faculty of Dentistry, Ordu University, Ordu, Turkey
| | - Ceren Gökmenoğlu
- Department of Periodontology, Faculty of Dentistry, Ordu University, Ordu, Turkey
| | - Emre Taha Deveci
- Department of Periodontology, Faculty of Dentistry, Ordu University, Ordu, Turkey
| | - Selman Çelen
- Department of Periodontology, Faculty of Dentistry, Ordu University, Ordu, Turkey
| | - Bahattin Avci
- Department of Medical Biochemistry, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Cankat Kara
- Department of Periodontology, Faculty of Dentistry, Ordu University, Ordu, Turkey
| | - Varol Çanakçi
- Department of Periodontology, Faculty of Dentistry, Ordu University, Ordu, Turkey
| |
Collapse
|
33
|
Andrade CA, Pacheco GA, Gálvez NMS, Soto JA, Bueno SM, Kalergis AM. Innate Immune Components that Regulate the Pathogenesis and Resolution of hRSV and hMPV Infections. Viruses 2020; 12:E637. [PMID: 32545470 PMCID: PMC7354512 DOI: 10.3390/v12060637] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
The human respiratory syncytial virus (hRSV) and human Metapneumovirus (hMPV) are two of the leading etiological agents of acute lower respiratory tract infections, which constitute the main cause of mortality in infants. However, there are currently approved vaccines for neither hRSV nor hMPV. Moreover, despite the similarity between the pathology caused by both viruses, the immune response elicited by the host is different in each case. In this review, we discuss how dendritic cells, alveolar macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid cells, and the complement system regulate both pathogenesis and the resolution of hRSV and hMPV infections. The roles that these cells play during infections by either of these viruses will help us to better understand the illnesses they cause. We also discuss several controversial findings, relative to some of these innate immune components. To better understand the inflammation in the lungs, the role of the respiratory epithelium in the recruitment of innate immune cells is briefly discussed. Finally, we review the main prophylactic strategies and current vaccine candidates against both hRSV and hMPV.
Collapse
Affiliation(s)
- Catalina A. Andrade
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Gaspar A. Pacheco
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Nicolas M. S. Gálvez
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Jorge A. Soto
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Susan M. Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| |
Collapse
|
34
|
Inhibition of IL-32 Expression Ameliorates Cerebral Ischemia-Reperfusion Injury via the NOD/MAPK/NF-κB Signaling Pathway. J Mol Neurosci 2020; 70:1713-1727. [PMID: 32474900 DOI: 10.1007/s12031-020-01557-0] [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: 02/19/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
Cerebral ischemia represents a major cause of disability, yet its precise mechanism remains unknown. In addition, ischemia-reperfusion injury which occurs during the blood recovery process increases the risk of mortality, and is not adequately addressed with current treatment. To improve therapeutic options, it is important to explore the vital substances that play a pivotal role in ischemia-reperfusion injury. This study is the first to investigate the role of IL-32, a vital pro-inflammatory factor, in models of cerebral ischemia-reperfusion injury. The results showed that IL-32 was highly expressed in both in vivo and in vitro models. The proteins of the NOD/MAPK/NF-κB pathway were also up-regulated, indicating a potential signaling pathway mechanism. Inhibition of IL-32 and blocking of the NOD/MAPK/NF-κB pathway increased cell survival, decreased the level of inflammatory factors and inflammasomes, and attenuated nitrosative stress. Taken together, the results show that inhibition of IL-32 expression ameliorates cerebral ischemia-reperfusion injury via the NOD/MAPK/NF-κB signaling pathway. The findings in this study reveal that IL-32 is a vital target of ischemia-reperfusion injury, providing a new avenue for treatment development.
Collapse
|
35
|
Cvitas I, Oberhänsli S, Leeb T, Dettwiler M, Müller E, Bruggman R, Marti EI. Investigating the epithelial barrier and immune signatures in the pathogenesis of equine insect bite hypersensitivity. PLoS One 2020; 15:e0232189. [PMID: 32343720 PMCID: PMC7188278 DOI: 10.1371/journal.pone.0232189] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/08/2020] [Indexed: 12/05/2022] Open
Abstract
Insect bite hypersensitivity (IBH) is a Th-2, IgE-mediated dermatitis of horses caused by bites of insects of the genus Culicoides that has common features with human atopic dermatitis. Together with Th-2 cells, the epithelial barrier plays an important role in development of type I hypersensitivities. In order to elucidate the role of the epithelial barrier and of the skin immune response in IBH we studied the transcriptome of lesional whole skin of IBH-horses (IBH-LE; n = 9) in comparison to non-lesional skin (IBH-NL; n = 8) as well as to skin of healthy control horses (H; n = 9). To study the "baseline state" of the epithelial barrier, we investigated the transcriptome of non-lesional epidermis in IBH-horses (EPI-IBH-NL; n = 10) in comparison with healthy epidermis from controls (EPI-H; n = 9). IBH-LE skin displayed substantial transcriptomic difference compared to H. IBH-LE was characterized by a downregulation of genes involved in tight junction formation, alterations in keratins and substantial immune signature of both Th-1 and Th-2 types with particular upregulation of IL13, as well as involvement of the hypoxic pathway. IBH-NL shared a number of differentially expressed genes (DEGs) with IBH-LE, but was overall more similar to H skin. In the epidermis, genes involved in metabolism of epidermal lipids, pruritus development, as well as IL25, were significantly differentially expressed between EPI-IBH-NL and EPI-H. Taken together, our data suggests an impairment of the epithelial barrier in IBH-affected horses that may act as a predisposing factor for IBH development. Moreover, these new mechanisms could potentially be used as future therapeutic targets. Importantly, many transcriptional features of equine IBH skin are shared with human atopic dermatitis, confirming equine IBH as a natural model of skin allergy.
Collapse
Affiliation(s)
- Iva Cvitas
- Division of Experimental Clinical Research, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- * E-mail:
| | - Simone Oberhänsli
- Interfaculty Bioinformatics Unit and SIB Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Tosso Leeb
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Genetics, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Martina Dettwiler
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Animal Pathology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Eliane Müller
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Animal Pathology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Department of Biomedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Remy Bruggman
- Interfaculty Bioinformatics Unit and SIB Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Eliane Isabelle Marti
- Division of Experimental Clinical Research, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
36
|
The Possible Anti-Inflammatory Effect of Dehydrocostus Lactone on DSS-Induced Colitis in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:5659738. [PMID: 32082397 PMCID: PMC7011397 DOI: 10.1155/2020/5659738] [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: 05/12/2019] [Revised: 07/18/2019] [Accepted: 08/01/2019] [Indexed: 01/07/2023]
Abstract
Background Dehydrocostus lactone (DL), one of the main active constituents in Aucklandia lappa Decne. (Muxiang), reported to have anti-inflammatory, antiulcer, and immunomodulatory properties. However, the effect of DL on ulcerative colitis (UC) has not been reported. To analyze the anti-inflammatory potential role of DL in UC, we provide a mechanism for the pharmacological action of DL. Methods The experimental model of UC was induced by using oral administration of 2% dextran sulfate sodium (DSS) with drinking water in BALB/c mice. Mesalazine (Mes, 0.52 g/kg/d), DL-high doses (DL-H, 20 mg/kg/d), DL-middle doses (DL-M, 15 mg/kg/d), DL-low doses (DL-L, 10 mg/kg/d) were gavaged once a day from day 4 to day 17. Disease activity index (DAI) was calculated daily. On day 18, mice were rapidly dissected and the colorectal tissues were used to detect the levels of UC-related inflammatory cytokines (TNF-α, IL-1β, MCP-1, MPO, SOD, IL-6, IL-17, and IL-23), IL-6/STAT3 inflammatory signaling pathway (iNOS, COX2, IL-6, GP130, L-17, and IL-23), and colorectal mucosal barrier-related regulatory factors (MUC2, XBP1s, and α, IL-1 Results DL reduced the colorectal inflammation histological assessment, decreased UC-related inflammatory cytokines (TNF-α, IL-1β, MCP-1, MPO, SOD, IL-6, IL-17, and IL-23), IL-6/STAT3 inflammatory signaling pathway (iNOS, COX2, IL-6, GP130, L-17, and IL-23), and colorectal mucosal barrier-related regulatory factors (MUC2, XBP1s, and α, IL-1 Conclusions DL possessed the potential of anti-inflammatory effect to treated colitis. The protective mechanism of DL may involve in reducing inflammation and improving colorectal barrier function via downregulating the IL-6/STAT3 signaling.
Collapse
|
37
|
Wang G, Ning FY, Wang JH, Yan HM, Kong HW, Zhang YT, Shen Q. Expression of interleukin-32 in bone marrow of patients with myeloma and its prognostic significance. World J Clin Cases 2019; 7:4234-4244. [PMID: 31911904 PMCID: PMC6940335 DOI: 10.12998/wjcc.v7.i24.4234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/14/2019] [Accepted: 11/26/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The guiding effect of prognostic stratification in multiple myeloma (MM) for treatment has been increasingly emphasized in recent years. The stratification of risk factors based on the International Staging System (ISS), Durie-Salmon (DS) staging and related indicators is affected by the renal function of patients, resulting in poor performance. This study assesses the relationship between interleukin-32 (IL-32) and related risk factors in 67 patients with MM and their clinical outcomes.
AIM To investigate the feasibility of IL-32 in evaluating prognosis in patients with MM and the factors influencing prognosis.
METHODS This was a pragmatic, prospective observational study of patients with MM at a single center. According to IL-32 level, patients were divided into two groups. The variables under consideration included age, blood β2-microglobulin, albumin, C-reactive protein, serum calcium, serum creatinine, lactate dehydrogenase, M protein type, ISS stage, DS stage, and IL-32 levels and minimal residual disease (MRD) after induction treatment. The main outcomes were progression-free survival (PFS) and overall survival (OS).
RESULTS IL-32 was an important factor affecting PFS and OS in patients with MM. Compared with patients with IL-32 levels ≥ 856.4 pg/mL, patients with IL-32 levels < 856.4 pg/mL had longer PFS (P = 0.0387) and OS (P = 0.0379); Univariate analysis showed that IL-32 level and MRD were significantly associated with OS and PFS (P < 0.05). Multivariate analysis showed that IL-32 levels ≥ 856.4 pg/mL and MRD positive were still independent risk factors for OS and PFS (P < 0.05).
CONCLUSION IL-32 is valuable for assessing the prognosis of MM patients. IL-32 level combined with MRD may be a useful routine evaluation index for MM patients after treatment.
Collapse
Affiliation(s)
- Gang Wang
- Department of Hematology, Quzhou People’s Hospital, Quzhou 324000, Zhejiang Province, China
| | - Fang-Ying Ning
- Department of Hematology, People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Jia-Heng Wang
- Department of Hematology, Quzhou People’s Hospital, Quzhou 324000, Zhejiang Province, China
| | - Hai-Meng Yan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Hong-Wei Kong
- Department of Hematology, Quzhou People’s Hospital, Quzhou 324000, Zhejiang Province, China
| | - Yu-Ting Zhang
- Adicon Clinical Laboratories Inc., Hangzhou 310023, Zhejiang Province, China
| | - Qiang Shen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
38
|
Catalan-Dibene J, McIntyre LL, Zlotnik A. Interleukin 30 to Interleukin 40. J Interferon Cytokine Res 2019; 38:423-439. [PMID: 30328794 DOI: 10.1089/jir.2018.0089] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cytokines are important molecules that regulate the ontogeny and function of the immune system. They are small secreted proteins usually produced upon activation of cells of the immune system, including lymphocytes and myeloid cells. Many cytokines have been described, and several have been recognized as pivotal players in immune responses and in human disease. In fact, several anticytokine antibodies have proven effective therapeutics, especially in various autoimmune diseases. In the last 15 years, new cytokines have been described, and many remain poorly understood. Among the most recent cytokines discovered are interleukins-30 (IL-30) to IL-40. Several of these are members of other cytokine superfamilies, including several IL-1 superfamily members (IL-33, IL-36, IL-37, and IL-38) as well as several new members of the IL-12 family (IL-30, IL-35, and IL-39). The rest (IL-31, IL-32, IL-34, and IL-40) are encoded by genes that do not belong to any cytokine superfamily. Our aim of this review was to present a concise version of the information available on these novel cytokines to facilitate their understanding by members of the immunological community.
Collapse
Affiliation(s)
- Jovani Catalan-Dibene
- 1 Department of Physiology and Biophysics and University of California , Irvine, Irvine, California.,2 Institute for Immunology, University of California, Irvine, Irvine, California
| | - Laura L McIntyre
- 3 Department of Molecular Biology and Biochemistry, University of California , Irvine, Irvine, California.,2 Institute for Immunology, University of California, Irvine, Irvine, California
| | - Albert Zlotnik
- 1 Department of Physiology and Biophysics and University of California , Irvine, Irvine, California.,2 Institute for Immunology, University of California, Irvine, Irvine, California
| |
Collapse
|
39
|
Yang Z, Shi L, Xue Y, Zeng T, Shi Y, Lin Y, Liu L. Interleukin-32 increases in coronary arteries and plasma from patients with coronary artery disease. Clin Chim Acta 2019; 497:104-109. [PMID: 31326419 DOI: 10.1016/j.cca.2019.07.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Interleukin-32 (IL-32) is a cytokine associated with higher risk of cardiovascular diseases in inflammatory environments. This study aimed to investigate the IL-32 levels in coronary artery disease (CAD) patients. METHODS IL-32 expression in coronary arteries from both normal donors and CAD patients were analyzed. Plasma IL-32, IFN-γ and IL-17 levels in stable angina pectoris (SAP, n = 80) patients, unstable angina pectoris (UAP, n = 96) patients, acute myocardial infarction (AMI, n = 72) patients and patients exhibiting chest pain unrelated to coronary artery disease (NCAD, n = 72) were measured. Additionally, whether plasma IL-32 levels were independent correlated with the presence of CAD was analyzed. RESULTS IL-32 was high expressed in atherosclerotic plaques of CAD patients when compared with normal coronary arteries, and macrophages were the major sources of IL-32. Compared with the NCAD group, IL-32, IFN-γ and IL-17 levels were increased in the CAD group and gradually increased through the SAP, UAP and AMI groups. Plasma IL-32 levels were positively correlated with the Gensini score, IFN-γ levels and IL-17 levels in CAD patients. The results of linear regression showed that IL-32 was independently associated with the occurrence of CAD. CONCLUSION Both the coronary artery and circulating IL-32 levels were increased in CAD patients and IL-32 may be a marker of noninvasive diagnosis of CAD.
Collapse
Affiliation(s)
- Zicong Yang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Lei Shi
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Yan Xue
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Tao Zeng
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Ying Shi
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Yingzhong Lin
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China.
| | - Ling Liu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China.
| |
Collapse
|
40
|
Lee YS, Kim KC, Mongre RK, Kim JY, Kim YR, Choi DY, Song S, Yun J, Han SB, Yoon DY, Hong JT. IL-32γ suppresses lung cancer stem cell growth via inhibition of ITGAV-mediated STAT5 pathway. Cell Death Dis 2019; 10:506. [PMID: 31263095 PMCID: PMC6602938 DOI: 10.1038/s41419-019-1737-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/02/2019] [Accepted: 06/10/2019] [Indexed: 12/16/2022]
Abstract
The cancer stem cells (CSCs) are thought to be responsible for cancer initiation, recurrence, and metastasis via a multifactorial process. IL-32γ has been known to inhibit several tumor developments. However, the role of IL-32γ in CSCs is unknown. The role of IL-32γ on tumor development was assessed in IL-32γ transgenic (Tg) mice allograft and xenograft model. In the in vitro assay, we analyzed CSC growth and apoptosis in cells with IL-32γ overexpression by cell viability assay and tumor-sphere formation assay. In addition, expression of cell proliferation, apoptosis markers, and signaling molecules was determined by western blot analysis. IL-32γ suppressed CD133+ CSC-induced allograft model in IL-32γ Tg mice and xenograft model. Tumor-sphere formation and cell viability assay revealed a greater inhibition of CSC proliferation and antineoplastic activity of IL-32γ in CD133+ CSCs as compared with normal cancer cells. The inhibitory effects of IL-32γ on tumor development were associated with inhibition of the STAT5 pathway. In addition, inhibition of STAT5 increased cleavage of caspase-3, but suppressed CD133 expression and colony formation. Web-based gene network analysis showed that IL-32 is correlated with ITGAV, an integrin gene. Our result revealed that knockdown of ITGAV by siRNA inhibited the phosphorylation of STAT5. Moreover, we identified that ITGAV overexpression reversed the effect of IL-32γ on phosphorylation of STAT5 and the expression of CD133. Our results demonstrate that IL-32γ negatively regulates CD133+ CSC proliferation and tumor development and suggest that IL-32γ has great potential for use in the treatment of cancer progression.
Collapse
Affiliation(s)
- Yong Sun Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Ki Cheon Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Raj Kumar Mongre
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Ji Young Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Yu Ri Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Dong Young Choi
- College of Pharmacy, Yeungnam University, Daehak-Ro 280, Gyeongsan, Gyeongsangbuk, Republic of Korea
| | - Sukgil Song
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Do Young Yoon
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 28160, Republic of Korea.
| |
Collapse
|
41
|
Abstract
Interleukin-32 (IL-32) was originally identified in natural killer (NK) cells activated by IL-2 in 1992. Thus, it was named NK cell transcript 4 (NK4) because of its unknown function at that time. The function of IL-32 has been elucidated over the last decade. IL-32 is primarily considered to be a booster of inflammatory reactions because it is induced by pro-inflammatory cytokines and stimulates the production of those cytokines and vice versa. Therefore, many studies have been devoted to studying the roles of IL-32 in inflammation-associated cancers, including gastric, colon cancer, and hepatocellular carcinoma. At the same time, roles of IL-32 have also been discovered in other cancers. Collectively, IL-32 fosters the tumor progression by nuclear factor-κB (NF-κB)-mediated cytokines and metalloproteinase production, as well as stimulation of differentiation into immunosuppressive cell types in some cancer types. However, it is also able to induce tumor cell apoptosis and enhance NK and cytotoxic T cell sensitivity in other cancer types. In this review, we will address the function of each IL-32 isoform in different cancer types studied to date, and suggest further strategies to comprehensively elucidate the roles of IL-32 in a context-dependent manner.
Collapse
Affiliation(s)
- Sora Han
- Research Institute for Women's Health, Sookmyung Women's University, Seoul 04310, Korea
| | - Young Yang
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| |
Collapse
|
42
|
Ma NX, Yin JC, Chen G. Transcriptome Analysis of Small Molecule-Mediated Astrocyte-to-Neuron Reprogramming. Front Cell Dev Biol 2019; 7:82. [PMID: 31231645 PMCID: PMC6558402 DOI: 10.3389/fcell.2019.00082] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/01/2019] [Indexed: 12/21/2022] Open
Abstract
Chemical reprogramming of astrocytes into neurons represents a promising approach to regenerate new neurons for brain repair, but the underlying mechanisms driving this trans-differentiation process are not well understood. We have recently identified four small molecules – CHIR99021, DAPT, LDN193189, and SB431542 – that can efficiently reprogram cultured human fetal astrocytes into functional neurons. Here we employ the next generation of RNA-sequencing technology to investigate the transcriptome changes during the astrocyte-to-neuron (AtN) conversion process. We found that the four small molecules can rapidly activate the hedgehog signaling pathway while downregulating many glial genes such as FN1 and MYL9 within 24 h of treatment. Chemical reprogramming is mediated by several waves of differential gene expression, including upregulation of hedgehog, Wnt/β-catenin, and Notch signaling pathways, together with downregulation of TGF-β and JAK/STAT signaling pathways. Our gene network analyses reveal many well-connected hub genes such as repulsive guidance molecule A (RGMA), neuronatin (NNAT), neurogenin 2 (NEUROG2), NPTX2, MOXD1, JAG1, and GAP43, which may coordinate the chemical reprogramming process. Together, these findings provide critical insights into the molecular cascades triggered by a combination of small molecules that eventually leads to chemical conversion of astrocytes into neurons.
Collapse
Affiliation(s)
- Ning-Xin Ma
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Jiu-Chao Yin
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Gong Chen
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
43
|
Meng Y, Wu T, Billings R, Kopycka-Kedzierawski DT, Xiao J. Human genes influence the interaction between Streptococcus mutans and host caries susceptibility: a genome-wide association study in children with primary dentition. Int J Oral Sci 2019; 11:19. [PMID: 31148553 PMCID: PMC6544625 DOI: 10.1038/s41368-019-0051-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/20/2019] [Accepted: 04/16/2019] [Indexed: 01/16/2023] Open
Abstract
Streptococcus mutans is a well-known cause of dental caries, due to its acidogenicity, aciduricity, and ability to synthesize exopolysaccharides in dental plaques. Intriguingly, not all children who carry S. mutans manifest caries, even with similar characteristics in oral hygiene, diet, and other environmental factors. This phenomenon suggests that host susceptibility potentially plays a role in the development of dental caries; however, the association between host genetics, S. mutans, and dental caries remains unclear. Therefore, this study examined the influence of host gene-by-S. mutans interaction on dental caries. Genome-wide association analyses were conducted in 709 US children (<13 years old), using the dbGap database acquired from the center for oral health research in appalachia (COHRA) and the Iowa Head Start programmes (GEIRS). A generalized estimating equation was used to examine the gene-by-S. mutans interaction effects on the outcomes (decayed and missing/filled primary teeth due to caries). Sequentially, the COHRA and GEIRS data were used to identify potential interactions and replicate the findings. Three loci at the genes interleukin 32 (IL32), galactokinase 2 (GALK2), and CUGBP, Elav-like family member 4 (CELF4) were linked to S. mutans carriage, and there was a severity of caries at a suggestive significance level among COHRA children (P < 9 × 10-5), and at a nominal significance level among GEIRS children (P = 0.047-0.001). The genetic risk score that combined the three loci also significantly interacted with S. mutans (P < 0.000 1). Functional analyses indicated that the identified genes are involved in the host immune response, galactose carbohydrate metabolism, and food-rewarding system, which could potentially be used to identify children at high risk for caries and to develop personalized caries prevention strategies.
Collapse
Affiliation(s)
- Ying Meng
- School of Nursing, University of Rochester, Rochester, NY, USA
| | - Tongtong Wu
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Ronald Billings
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Jin Xiao
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
44
|
Paz H, Tsoi J, Kalbasi A, Grasso CS, McBride WH, Schaue D, Butterfield LH, Maurer DM, Ribas A, Graeber TG, Economou JS. Interleukin 32 expression in human melanoma. J Transl Med 2019; 17:113. [PMID: 30953519 PMCID: PMC6449995 DOI: 10.1186/s12967-019-1862-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023] Open
Abstract
Background Various proinflammatory cytokines can be detected within the melanoma tumor microenvironment. Interleukin 32 (IL32) is produced by T cells, NK cells and monocytes/macrophages, but also by a subset of melanoma cells. We sought to better understand the biology of IL32 in human melanoma. Methods We analyzed RNA sequencing data from 53 in-house established human melanoma cell lines and 479 melanoma tumors from The Cancer Genome Atlas dataset. We evaluated global gene expression patterns associated with IL32 expression. We also evaluated the impact of proinflammatory molecules TNFα and IFNγ on IL32 expression and dedifferentiation in melanoma cell lines in vitro. In order to study the transcriptional regulation of IL32 in these cell lines, we cloned up to 10.5 kb of the 5′ upstream region of the human IL32 gene into a luciferase reporter vector. Results A significant proportion of established human melanoma cell lines express IL32, with its expression being highly correlated with a dedifferentiation genetic signature (high AXL/low MITF). Non IL32-expressing differentiated melanoma cell lines exposed to TNFα or IFNγ can be induced to express the three predominant isoforms (α, β, γ) of IL32. Cis-acting elements within this 5′ upstream region of the human IL32 gene appear to govern both induced and constitutive gene expression. In the tumor microenvironment, IL32 expression is highly correlated with genes related to T cell infiltration, and also positively correlates with high AXL/low MITF dedifferentiated gene signature. Conclusions Expression of IL32 in human melanoma can be induced by TNFα or IFNγ and correlates with a treatment-resistant dedifferentiated genetic signature. Constitutive and induced expression are regulated, in part, by cis-acting sequences within the 5′ upstream region. Electronic supplementary material The online version of this article (10.1186/s12967-019-1862-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Helicia Paz
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Jennifer Tsoi
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA, 90095, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Anusha Kalbasi
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA.,Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - Catherine S Grasso
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - William H McBride
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Lisa H Butterfield
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA.,Department of Surgery, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA.,Department of Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Deena M Maurer
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Antoni Ribas
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA, 90095, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - James S Economou
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA. .,Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA. .,Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA, 90095, USA. .,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
45
|
Shin JM, Kim HJ, Park JH, Hwang YJ, Lee HM. Asian Sand Dust Regulates IL-32 Production in Airway Epithelial Cells: Inhibitory Effect of Glucocorticoids. Am J Rhinol Allergy 2019; 33:403-412. [PMID: 30919652 DOI: 10.1177/1945892419839538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose Epidemiologic studies have reported that Asian sand dust (ASD) is associated with chronic inflammatory diseases of the respiratory system. Glucocorticoids (GCs) have potent anti-inflammatory properties. The aims of this study were to evaluate the effects of GCs on ASD-induced interleukin-32 (IL-32) expression and to identify the underlying signaling pathways in airway epithelial cells. Methods A 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was used to evaluate cytotoxicity in A549 and human primary nasal epithelial cells. Expression levels of IL-32 messenger RNA and protein were measured by Western blot, real-time polymerase chain reaction, ELISA, and immunofluorescence staining. Signaling pathways were analyzed using specific inhibitors of Akt, MAPK, or NF- κB. The effects of GCs on the expression of ASD-induced IL-32 were confirmed with ex vivo organ cultures of the nasal interior turbinate. Results ASD (0–400 ng/mL) had no significant cytotoxic effects in A549 cells and human primary nasal epithelial cells. Expression levels of IL-32 were dose-dependently upregulated by ASD treatment in A549 cells. ASD induced phosphorylation of Akt, MAPK, and NF-κB, whereas GCs and specific inhibitors of Akt, MAPK, and NF-κB downregulated these activations and the expression of IL-32. These findings were further confirmed in human primary nasal epithelial cells and ex vivo organ cultures of the nasal interior turbinate. Conclusions GCs have an inhibitory effect on ASD-induced IL-32 expression via the Akt, MAPK, and NF- κB signaling pathways in airway epithelial cells.
Collapse
Affiliation(s)
- Jae-Min Shin
- 1 Department of Otorhinolaryngology - Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hwee-Jin Kim
- 2 Division of Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Joo-Hoo Park
- 2 Division of Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - You Jin Hwang
- 3 Department of Life Science, College of BioNano, Gachon University, Incheon, Republic of Korea
| | - Heung-Man Lee
- 1 Department of Otorhinolaryngology - Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea.,2 Division of Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea.,4 Institute for Korea University IVD Support Center, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
46
|
Santinelli L, Statzu M, Pierangeli A, Frasca F, Bressan A, Pinacchio C, Nonne C, Turriziani O, Antonelli G, d'Ettorre G, Scagnolari C. Increased expression of IL-32 correlates with IFN-γ, Th1 and Tc1 in virologically suppressed HIV-1-infected patients. Cytokine 2019; 120:273-281. [PMID: 30910260 DOI: 10.1016/j.cyto.2019.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 01/07/2019] [Accepted: 01/27/2019] [Indexed: 02/06/2023]
Abstract
Following recent attention focused on IL-32 as an important component involved in the inflammatory cytokine network, we speculated that IL-32's action on IFN-γ and IFN-γ secreting T cell subsets may help sustain the immune activation and dysregulation found in patients with HIV-1 achieving viral suppression. To explore this hypothesis, transcript levels of IL-32 and IFN-γ were evaluated in PBMC from 139 virologically suppressed HIV-1-infected patients and from 63 healthy individuals by Real Time RT-PCR assays. IL-32 and IFN-γ mRNA levels were also analyzed in CD4+ T cells, CD14+ monocytes and lamina propria lymphocytes (LPL) of the gut district in a subgroup of HIV-1-infected subjects. IFN-γ secreting CD4+ (Th1) and CD8+ (Tc1) T cell subset frequencies were evaluated in LPL by multiparametric flow cytometry. Gene expression results revealed that IL-32 and IFN-γ levels in PBMC from HIV-1-positive patients were significantly elevated compared to those from healthy donors, correlated with each other and increased with patient age. Both IL-32 and IFN-γ genes were also more strongly expressed in CD4+ T cells than in CD14+ monocytes. By contrast, IL-32 levels in LPL were comparable to those measured in PBMC, while IFN-γ levels were higher in PBMC than those in LPL. Negative correlations were found between IL-32 levels and the frequencies of Th1 and Tc1 subsets in gut mucosa. Collectively, our results provide the first evidence that IL-32 levels remain elevated in treated HIV-1-infected patients and correlate with IFN-γ, Th1 and Tc1 subsets.
Collapse
Affiliation(s)
- Letizia Santinelli
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Maura Statzu
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Alessandra Pierangeli
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Federica Frasca
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Alessia Bressan
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy; Microbiology and Virology Unit, Sapienza University Hospital, Rome, Italy.
| | - Claudia Pinacchio
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.
| | - Chiara Nonne
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Ombretta Turriziani
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Guido Antonelli
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy; Microbiology and Virology Unit, Sapienza University Hospital, Rome, Italy.
| | - Gabriella d'Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.
| | - Carolina Scagnolari
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
47
|
Ebersole JL, Peyyala R, Gonzalez OA. Biofilm-induced profiles of immune response gene expression by oral epithelial cells. Mol Oral Microbiol 2019; 34. [PMID: 30407731 DOI: 10.1111/omi.12251] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2018] [Indexed: 12/12/2022]
Abstract
This study examined the oral epithelial immunotranscriptome response patterns modulated by oral bacterial planktonic or biofilm challenge. We assessed gene expression patterns when epithelial cells were challenged with a multispecies biofilm composed of Streptococcus gordonii, Fusobacterium nucleatum, and Porphyromonas gingivalis representing a type of periodontopathic biofilm compared to challenge with the same species of planktonic bacteria. Of the 579 human immunology genes, a substantial signal of the epithelial cells was observed to 181 genes. Biofilm challenged stimulated significant elevations compared to planktonic bacteria for IL32, IL8, CD44, B2M, TGFBI, NFKBIA, IL1B, CD59, IL1A, CCL20 representing the top 10 signals comprising 55% of the overall signal for the epithelial cell responses. Levels of PLAU, CD9, IFITM1, PLAUR, CD24, TNFSF10, and IL1RN were all elevated by each of the planktonic bacterial challenge vs the biofilm responses. While the biofilms up-regulated 123/579 genes (>2-fold), fewer genes were increased by the planktonic species (36 [S gordonii], 30 [F nucleatum], 44 [P gingivalis]). A wide array of immune genes were regulated by oral bacterial challenge of epithelial cells that would be linked to the local activity of innate and adaptive immune response components in the gingival tissues. Incorporating bacterial species into a structured biofilm dramatically altered the number and level of genes expressed. Additionally, a specific set of genes were significantly decreased with the multispecies biofilms suggesting that some epithelial cell biologic pathways are down-regulated when in contact with this type of pathogenic biofilm.
Collapse
Affiliation(s)
- Jeffrey L Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, Nevada.,College of Dentistry, Center for Oral Health Research, University of Kentucky, Lexington, Kentucky
| | - Rebecca Peyyala
- College of Dentistry, Center for Oral Health Research, University of Kentucky, Lexington, Kentucky
| | - Octavio A Gonzalez
- College of Dentistry, Center for Oral Health Research, University of Kentucky, Lexington, Kentucky.,Division of Periodontology, College of Dentistry, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
48
|
Neupane S, Srivastav S, Bhurtel S, Katila N, Shadfar S, Park PH, Hong JT, Choi DY. Enhanced neuroinflammatory responses after systemic LPS injection in IL-32β transgenic mice. J Chem Neuroanat 2018; 94:173-182. [DOI: 10.1016/j.jchemneu.2018.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022]
|
49
|
Zou GS, Li SJ, Zheng SL, Pan X, Huang ZP. Lemon-Peel extract ameliorates rheumatoid arthritis by reducing xanthine oxidase and inflammatory cytokine levels. J Taiwan Inst Chem Eng 2018. [DOI: 10.1016/j.jtice.2018.07.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
50
|
Lee YS, Lee CH, Bae JT, Nam KT, Moon DB, Hwang OK, Choi JS, Kim TH, Jun HO, Jung YS, Hwang DY, Han SB, Yoon DY, Hong JT. Inhibition of skin carcinogenesis by suppression of NF-κB dependent ITGAV and TIMP-1 expression in IL-32γ overexpressed condition. J Exp Clin Cancer Res 2018; 37:293. [PMID: 30486830 PMCID: PMC6263970 DOI: 10.1186/s13046-018-0943-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Interleukin-32 (IL-32) has been associated with various diseases. Previous studies have shown that IL-32 inhibited the development of several tumors. However, the role of IL-32γ, an isotype of IL-32, in skin carcinogenesis remains unknown. METHODS We compared 7,12-Dimethylbenz[a]anthracene/12-O-Tetradecanoylphorbol-13-acetate (DMBA/TPA)-induced skin carcinogenesis in wild type (WT) and IL-32γ-overexpressing mice to evaluate the role of IL-32γ. We also analyzed cancer stemness and NF-κB signaling in skin cancer cell lines with or without IL-32γ expression by western blotting, quantitative real-time PCR and immunohistochemistry analysis. RESULTS Carcinogen-induced tumor incidence in IL-32γ mice was significantly reduced in comparison to that in WT mice. Infiltration of inflammatory cells and the expression levels of pro-inflammatory mediators were decreased in the skin tumor tissues of IL-32γ mice compared with WT mice. Using a genome-wide association study analysis, we found that IL-32 was associated with integrin αV (ITGAV) and tissue inhibitor of metalloproteinase-1 (TIMP-1), which are critical factor for skin carcinogenesis. Reduced expression of ITGAV and TIMP-1 were identified in DMBA/TPA-induced skin tissues of IL-32γ mice compared to that in WT mice. NF-κB activity was also reduced in DMBA/TPA-induced skin tissues of IL-32γ mice. IL-32γ decreased cancer cell sphere formation and expression of stem cell markers, and increased chemotherapy-induced cancer cell death. IL-32γ also downregulated expression of ITGAV and TIMP-1, accompanied with the inhibition of NF-κB activity. In addition, IL-32γ expression with NF-κB inhibitor treatment further reduced skin inflammation, epidermal hyperplasia, and cancer cell sphere formation and downregulated expression levels of ITGAV and TIMP-1. CONCLUSIONS These findings indicated that IL-32γ suppressed skin carcinogenesis through the inhibition of both stemness and the inflammatory tumor microenvironment by the downregulation of TIMP-1 and ITGAV via inactivation of NF-κB signaling.
Collapse
Affiliation(s)
- Yong Sun Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Chung Hee Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
- Hanbul Co, Ltd. R&D center, 634 Eon Ju-Ro, Gangnam-gu, Seoul, Republic of Korea
| | - Jun Tae Bae
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Kyung Tak Nam
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Dae Bong Moon
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Ok Kyung Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Jeong Soon Choi
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Tae Hoon Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Hyoung Ok Jun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Young Suk Jung
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Dae Yeon Hwang
- Department of Biomaterial Science, Pusan National University, Miryang, Kyungnam 50463 Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Do Young Yoon
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Gwangjin-gu, Seoul, 05029 Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| |
Collapse
|