1
|
Mohanty C, Singh CK, Daccache JA, Damsky W, Kendziorski C, Yan D, Prasad A, Zhang D, Keenan T, Drolet B, Ahmad N, Shields BE. Granuloma Annulare Exhibits Mixed Immune and Macrophage Polarization Profiles with Spatial Transcriptomics. J Invest Dermatol 2025; 145:109-121. [PMID: 38844128 DOI: 10.1016/j.jid.2024.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/11/2024] [Accepted: 04/27/2024] [Indexed: 07/16/2024]
Abstract
Granuloma annulare (GA) is an idiopathic condition characterized by granulomatous inflammation in the skin. Prior studies have suggested that GA develops from various triggers, leading to a complex interplay involving innate and adaptive immunity, tissue remodeling, and fibrosis. Macrophages are the major immune cells comprising GA granulomas; however, the molecular drivers and inflammatory signaling cascade behind macrophage activation are poorly understood. Histologically, GA exhibits both palisaded and interstitial patterns on histology; however, the molecular composition of GA at the spatial level remains unexplored. GA is a condition without Food and Drug Administration-approved therapies despite the significant impact of GA on QOL. Spatial transcriptomics is a valuable tool for profiling localized, genome-wide gene expression changes across tissues, with emerging applications in clinical medicine. To improve our understanding of the spatially localized gene expression patterns underlying GA, we profiled the spatial gene expression landscape from 6 patients with GA. Our findings revealed mixed T helper 1 and T helper 2 signals comprising the GA microenvironment and spatially distinct M1 and M2 macrophage polarization characteristics. IFN-γ and TNF signals emerged as important regulators of GA granulomatous inflammation, and IL-32 emerged as a key driver of granulomatous inflammation. Overall, our spatial transcriptomics data indicate that GA exhibits mixed immune and macrophage polarization.
Collapse
Affiliation(s)
- Chitrasen Mohanty
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Chandra K Singh
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joseph A Daccache
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Pathology, NYU Langone Health, New York, New York, USA
| | - William Damsky
- Department of Pathology, NYU Langone Health, New York, New York, USA; Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Di Yan
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Aman Prasad
- Department of Dermatology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Donglin Zhang
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Tom Keenan
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Beth Drolet
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Nihal Ahmad
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Dermatology, William S. Middleton Memorial Veterans' Hospital, Madison, Wisconsin, USA
| | - Bridget E Shields
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
2
|
Yin QZ, Liu YJ, Zhang Q, Xi SY, Yang TB, Li JP, Gao J. Overexpression of Basonuclin Zinc Finger Protein 2 in stromal cell is related to mesenchymal phenotype and immunosuppression of mucinous colorectal adenocarcinoma. Int Immunopharmacol 2024; 142:113184. [PMID: 39306894 DOI: 10.1016/j.intimp.2024.113184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Mucinous carcinoma (MC) is a distinct histologic subtype of colorectal cancer (CRC) that is less studied and associated with poor prognosis. This study aimed to identify MC-specific therapeutic targets and biomarkers to improve the prognosis of this aggressive disease. METHODS CRC samples from The Cancer Genome Atlas (TCGA) were categorized into MC and non-MC (NMC) groups based on histologic type. A multi-scale embedded gene co-expression network analysis (MEGENA) was constructed to identify gene modules associated with the MC group. The potential functions of Basonuclin Zinc Finger Protein 2 (BNC2) were further analyzed using the Biomarker Exploration for Solid Tumors (BEST) database. In vivo and in vitro experiments were conducted to validate the predicted results. RESULTS We identified the stromal component-related gene, BNC2, in the MC population. This gene is associated with a shorter progression-free interval (PFI) in CRC patients. BNC2 promotes FAP (encoding Fibroblast Activation Protein Alpha) transcription in cancer-associated fibroblasts (CAFs) and is involved in angiogenesis through two pathways. Additionally, BNC2 enhances tumor cell invasiveness in a CAF-dependent manner. Patients with high BNC2 expression benefited less from immunotherapy compared to those with low BNC2 expression. CONCLUSIONS Our study highlights the clinical importance of BNC2 in MC, and targeting BNC2 on stromal cells (fibroblasts and endothelial cells) may be an effective strategy for treating MC.
Collapse
Affiliation(s)
- Qing-Zhong Yin
- Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yuan-Jie Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Qian Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Song-Yang Xi
- Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, Jiangsu 212000, China
| | - Tian-Bao Yang
- Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jie-Pin Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| | - Ju Gao
- The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, Jiangsu 225009, China; Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
3
|
Ramani H, Gosselin A, Bunet R, Jenabian MA, Sylla M, Pagliuzza A, Chartrand-Lefebvre C, Routy JP, Goulet JP, Thomas R, Trottier B, Martel-Laferrière V, Fortin C, Chomont N, Fromentin R, Landay AL, Durand M, Ancuta P, El-Far M, Tremblay C. IL-32 Drives the Differentiation of Cardiotropic CD4+ T Cells Carrying HIV DNA in People With HIV. J Infect Dis 2024; 229:1277-1289. [PMID: 38113908 PMCID: PMC11095560 DOI: 10.1093/infdis/jiad576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023] Open
Abstract
Interleukin 32 (IL-32) is a potent multi-isoform proinflammatory cytokine, which is upregulated in people with HIV (PWH) and is associated with cardiovascular disease (CVD) risk. However, the impact of IL-32 isoforms on CD4 T-cell cardiotropism, a mechanism potentially contributing to heart inflammation, remains unknown. Here we show that IL-32 isoforms β and γ induce the generation of CCR4+CXCR3+ double positive (DP) memory CD4 T-cell subpopulation expressing the tyrosine kinase receptor c-Met, a phenotype associated with heart-homing of T cells. Our ex vivo studies on PWH show that the frequency of DP CD4 T cells is significantly higher in individuals with, compared to individuals without, subclinical atherosclerosis and that DP cells from antiretroviral-naive and treated individuals are highly enriched with HIV DNA. Together, these data demonstrate that IL-32 isoforms have the potential to induce heart-homing of HIV-infected CD4 T cells, which may further aggravate heart inflammation and CVD in PWH.
Collapse
Affiliation(s)
- Hardik Ramani
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Annie Gosselin
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Rémi Bunet
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Mohammad-Ali Jenabian
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Department of Biological Sciences, Université du Québec Montréal, Montréal, Québec, Canada
| | - Mohamed Sylla
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Amélie Pagliuzza
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Carl Chartrand-Lefebvre
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Département de Radiologie, Radio-oncologie et Médecine Nucléaire, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Pierre Routy
- Research Institute, McGill University Health Centre, Montréal, Québec, Canada
| | | | - Réjean Thomas
- Clinique Médicale l’Actuel, Montréal, Québec, Canada
| | - Benoit Trottier
- Clinique de Médecine Urbaine du Quartier Latin, Montréal, Québec, Canada
| | - Valérie Martel-Laferrière
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Claude Fortin
- Department of Medical Microbiology and Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Nicolas Chomont
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Rémi Fromentin
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Alan L Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Madeleine Durand
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Petronela Ancuta
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Mohamed El-Far
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Cecile Tremblay
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| |
Collapse
|
4
|
Costa TFR, Catta-Preta CMC, Goundry A, Carvalho DB, Rodrigues NS, Vivarini AC, de Abreu MF, Reis FCG, Lima APCA. The ecotin-like peptidase inhibitor of Trypanosoma cruzi prevents TMPRSS2-PAR2-TLR4 crosstalk downmodulating infection and inflammation. FASEB J 2024; 38:e23566. [PMID: 38526868 DOI: 10.1096/fj.202302091rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/24/2024] [Accepted: 03/06/2024] [Indexed: 03/27/2024]
Abstract
Trypanosoma cruzi is the causative agent of Chagas disease, a chronic pathology that affects the heart and/or digestive system. This parasite invades and multiplies in virtually all nucleated cells, using a variety of host cell receptors for infection. T. cruzi has a gene that encodes an ecotin-like inhibitor of serine peptidases, ISP2. We generated ISP2-null mutants (Δisp2) in T. cruzi Dm28c using CRISPR/Cas9. Epimastigotes of Δisp2 grew normally in vitro but were more susceptible to lysis by human serum compared to parental and ISP2 add-back lines. Tissue culture trypomastigotes of Δisp2 were more infective to human muscle cells in vitro, which was reverted by the serine peptidase inhibitors aprotinin and camostat, suggesting that host cell epitheliasin/TMPRSS2 is the target of ISP2. Pretreatment of host cells with an antagonist to the protease-activated receptor 2 (PAR2) or an inhibitor of Toll-like receptor 4 (TLR4) selectively counteracted the increased cell invasion by Δisp2, but did not affect invasion by parental and add-back lines. The same was observed following targeted gene silencing of PAR2, TLR4 or TMPRSS2 in host cells by siRNA. Furthermore, Δisp2 caused increased tissue edema in a BALB/c mouse footpad infection model after 3 h differently to that observed following infection with parental and add-back lines. We propose that ISP2 contributes to protect T. cruzi from the anti-microbial effects of human serum and to prevent triggering of PAR2 and TLR4 in host cells, resulting in the modulation of host cell invasion and contributing to decrease inflammation during acute infection.
Collapse
Affiliation(s)
- Tatiana F R Costa
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina M C Catta-Preta
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amy Goundry
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle B Carvalho
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nathalia S Rodrigues
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aislan C Vivarini
- Departamento de Biologia Celular e Molecular, Insituto de Biologia, Universidade Federal Fluminense, Niteroi, Brazil
| | - Mayra Fonseca de Abreu
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavia C G Reis
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula C A Lima
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Furuya K, Nakajima M, Tsunedomi R, Nakagami Y, Xu M, Matsui H, Tokumitsu Y, Shindo Y, Watanabe Y, Tomochika S, Maeda N, Iida M, Suzuki N, Takeda S, Hazama S, Ioka T, Hoshii Y, Ueno T, Nagano H. High serum proteinase-3 levels predict poor progression-free survival and lower efficacy of bevacizumab in metastatic colorectal cancer. BMC Cancer 2024; 24:165. [PMID: 38308214 PMCID: PMC10835931 DOI: 10.1186/s12885-024-11924-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/26/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND To improve the prognosis of patients with metastatic colorectal cancer (mCRC), investigating predictive biomarkers of their prognosis and chemotherapeutic responsiveness is necessary. This study aimed to analyze the clinical significance of serum proteinase-3 (PRTN3) as a predictor for prognosis and chemosensitivity, especially to bevacizumab therapy, in mCRC. METHODS This single-center retrospective observational study enrolled 79 patients with mCRC in our hospital and 353 patients with colorectal cancer in the TCGA database. Preoperative serum PRTN3 levels were measured using an enzyme-linked immunosorbent assay. The clinicopathological characteristics and prognosis according to serum PRTN3 levels were then evaluated. PRTN3 expression in tumor and stromal cells was evaluated immunohistochemically. The impact of PRTN3 levels on angiogenesis and bevacizumab sensitivity was evaluated using the tube formation assay. RESULTS Serum PRTN3 levels were an independent poor prognostic factor for progression-free survival (PFS) (hazard ratio, 2.082; 95% confidence interval, 1.118-3.647; P=0.010) in patients with mCRC. Similarly, prognostic analysis with TCGA data sets showed poorer overall survival in patients with PRTN3 expression than that in patients without PRTN3 expression, especially in patients with stage IV. Immunohistochemical analysis of resected specimens revealed that stromal neutrophils expressed PRTN3, and their expression level was significantly correlated with serum PRTN3 levels. Interestingly, the effectiveness of first-line chemotherapy was significantly poorer in the high serum PRTN3 level group. High serum PRTN3 was significantly associated with poor PFS (hazard ratio, 3.027; 95% confidence interval, 1.175-7.793; P=0.0161) in patients treated with bevacizumab, an anti-angiogenic inhibitor. The tube formation assay revealed that PRTN3 administration notably augmented angiogenesis while simultaneously attenuating the anti-angiogenic influence exerted by bevacizumab therapy. CONCLUSIONS Serum PRTN3 levels could be a novel predictive biomarker of PFS of first-line chemotherapy, especially for bevacizumab therapy, in patients with mCRC.
Collapse
Affiliation(s)
- Kei Furuya
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Masao Nakajima
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yuki Nakagami
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Ming Xu
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Hiroto Matsui
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yukio Tokumitsu
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yoshitaro Shindo
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yusaku Watanabe
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shinobu Tomochika
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Noriko Maeda
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Michihisa Iida
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shigeru Takeda
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shoichi Hazama
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Tatsuya Ioka
- Oncology Center, Yamaguchi University Hospital, Ube, Yamaguchi, 755-8505, Japan
| | - Yoshinobu Hoshii
- Department of Diagnostic Pathology, Yamaguchi University Hospital, Ube, Yamaguchi, 755-8505, Japan
| | - Tomio Ueno
- Department of Digestive Surgery, Kawasaki Medical School, Kurashiki, Okayama, 701-0192, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| |
Collapse
|
6
|
Sasidharan K, Caddeo A, Jamialahmadi O, Noto FR, Tomasi M, Malvestiti F, Ciociola E, Tavaglione F, Mancina RM, Cherubini A, Bianco C, Mirarchi A, Männistö V, Pihlajamäki J, Kärjä V, Grimaudo S, Luukkonen PK, Qadri S, Yki-Järvinen H, Petta S, Manfrini S, Vespasiani-Gentilucci U, Bruni V, Valenti L, Romeo S. IL32 downregulation lowers triglycerides and type I collagen in di-lineage human primary liver organoids. Cell Rep Med 2024; 5:101352. [PMID: 38232700 PMCID: PMC10829727 DOI: 10.1016/j.xcrm.2023.101352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 09/26/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024]
Abstract
Steatotic liver disease (SLD) prevails as the most common chronic liver disease yet lack approved treatments due to incomplete understanding of pathogenesis. Recently, elevated hepatic and circulating interleukin 32 (IL-32) levels were found in individuals with severe SLD. However, the mechanistic link between IL-32 and intracellular triglyceride metabolism remains to be elucidated. We demonstrate in vitro that incubation with IL-32β protein leads to an increase in intracellular triglyceride synthesis, while downregulation of IL32 by small interfering RNA leads to lower triglyceride synthesis and secretion in organoids from human primary hepatocytes. This reduction requires the upregulation of Phospholipase A2 group IIA (PLA2G2A). Furthermore, downregulation of IL32 results in lower intracellular type I collagen levels in di-lineage human primary hepatic organoids. Finally, we identify a genetic variant of IL32 (rs76580947) associated with lower circulating IL-32 and protection against SLD measured by non-invasive tests. These data suggest that IL32 downregulation may be beneficial against SLD.
Collapse
Affiliation(s)
- Kavitha Sasidharan
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Andrea Caddeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Francesca Rita Noto
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Melissa Tomasi
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Malvestiti
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Ester Ciociola
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Federica Tavaglione
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Rosellina M Mancina
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Alessandro Cherubini
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cristiana Bianco
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Angela Mirarchi
- Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Clinical Nutrition and Obesity Centre, Kuopio University Hospital, Kuopio, Finland
| | - Vesa Kärjä
- Department of Pathology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Stefania Grimaudo
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Palermo, Italy
| | - Panu K Luukkonen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Sami Qadri
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Palermo, Italy
| | - Silvia Manfrini
- Operative Unit of Endocrinology and Diabetes, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Research Unit of Endocrinology and Diabetes, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Umberto Vespasiani-Gentilucci
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Vincenzo Bruni
- Operative Unit of Bariatric Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Luca Valenti
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy.
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy; Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
7
|
Wallimann A, Schenk M. IL-32 as a potential biomarker and therapeutic target in skin inflammation. Front Immunol 2023; 14:1264236. [PMID: 37727785 PMCID: PMC10505650 DOI: 10.3389/fimmu.2023.1264236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/15/2023] [Indexed: 09/21/2023] Open
Abstract
IL-32 is a recently described cytokine that performs a variety of functions under inflammatory conditions. Serum IL-32 has been shown to be elevated in several diseases, including type 2 diabetes, cancer, systemic lupus erythematosus, HIV infection, and atopic diseases including atopic dermatitis. There are nine different isoforms of IL-32, with IL-32γ being the most biologically active one. The following review summarizes the different roles of the various IL-32 isoforms in the context of skin inflammation, with a focus on atopic dermatitis.
Collapse
Affiliation(s)
- Alexandra Wallimann
- Christine Kühne – Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Mirjam Schenk
- Christine Kühne – Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Institute of Tissue Medicine and Pathology, Experimental Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Chettab K, Fitzsimmons C, Novikov A, Denis M, Phelip C, Mathé D, Choffour PA, Beaumel S, Fourmaux E, Norca P, Kryza D, Evesque A, Jordheim LP, Perrial E, Matera EL, Caroff M, Kerzerho J, Dumontet C. A systemically administered detoxified TLR4 agonist displays potent antitumor activity and an acceptable tolerance profile in preclinical models. Front Immunol 2023; 14:1066402. [PMID: 37223101 PMCID: PMC10200957 DOI: 10.3389/fimmu.2023.1066402] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/19/2023] [Indexed: 05/25/2023] Open
Abstract
Bacterial lipopolysaccharides (LPS) are potent innate immunostimulants targeting the Toll-like receptor 4 (TLR4), an attractive and validated target for immunostimulation in cancer therapy. Although LPS possess anti-tumor activity, toxicity issues prevent their systemic administration at effective doses in humans. We first demonstrated that LPS formulated in liposomes preserved a potent antitumor activity per se upon systemic administration in syngeneic models, and significantly enhance the antitumor activity of the anti-CD20 antibody rituximab in mice xenografted with the human RL lymphoma model. Liposomal encapsulation also allowed a 2-fold reduction in the induction of pro-inflammatory cytokines by LPS. Mice receiving an intravenous administration demonstrated a significant increase of neutrophils, monocytes and macrophages at the tumor site as well as an increase of macrophages in spleen. Further, we chemically detoxified LPS to obtain MP-LPS that was associated with a 200-fold decrease in the induction of proinflammatory cytokines. When encapsulated in a clinically approved liposomal formulation, toxicity, notably pyrogenicity (10-fold), was limited while the antitumor activity and immunoadjuvant effect were maintained. This improved tolerance profile of liposomal MP-LPS was associated with the preferential activation of the TLR4-TRIF pathway. Finally, in vitro studies demonstrated that stimulation with encapsulated MP-LPS reversed the polarization of M2 macrophages towards an M1 phenotype, and a phase 1 trial in healthy dogs validated its tolerance upon systemic administration up to very high doses (10µg/kg). Altogether, our results demonstrate the strong therapeutic potential of MPLPS formulated in liposomes as a systemically active anticancer agent, supporting its evaluation in patients with cancer.
Collapse
Affiliation(s)
- Kamel Chettab
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon, France
- Hospices Civils de Lyon, Lyon, France
| | - Chantel Fitzsimmons
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon, France
| | - Alexey Novikov
- HEPHAISTOS-Pharma, Université Paris-Saclay, Orsay, France
| | - Morgane Denis
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon, France
- Antinéo, Lyon, France
| | | | | | | | - Sabine Beaumel
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon, France
| | - Eric Fourmaux
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon, France
| | - Patrick Norca
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon, France
| | | | | | - Lars Petter Jordheim
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon, France
| | - Emeline Perrial
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon, France
| | - Eva-Laure Matera
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon, France
| | - Martine Caroff
- HEPHAISTOS-Pharma, Université Paris-Saclay, Orsay, France
| | | | - Charles Dumontet
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon, France
- Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
9
|
Singh H, Kumar U, Senapati S. Translational implications of humoral and cellular immune dysfunction in granulomatosis with polyangiitis. Cytokine 2023; 164:156154. [PMID: 36812668 DOI: 10.1016/j.cyto.2023.156154] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/11/2023] [Indexed: 02/22/2023]
Abstract
Granulomatosis with polyangiitis (GPA) is a rare systemic ANCA (Anti-neutrophil cytoplasmic antibodies) associated vasculitis (AAV). In the last couple of decades, GPA has emerged as a disease of concern due to rapid increase in the prevalence and incidence especially in developing countries. Unknown aetiology and rapid progression have made GPA a critical disease. Thus, establishing specific tools to facilitate early and faster disease diagnosis and efficient disease management has immense importance. GPA may develop in genetically predisposed individuals on receiving the external stimulus (i.e. microbial pathogen, pollutant etc.) that triggers the immune response. B-cell activating factor (BAFF) produced by the neutrophils, promotes the B-cell maturation and survival which leads to increased ANCA production. Abnormal B-cell and T-cell proliferation and their cytokine response plays a major role in disease pathogenesis and granuloma formation. ANCA interacts with neutrophils and induces the neutrophil extracellular traps (NETs) formation and reactive oxygen species (ROS) production which leads to the endothelial cell injury. This review article summarizes the critical pathological events and how cytokines and immune cells shape the GPA pathogenesis. Decoding this complex network would facilitate in developing tools for diagnosis, prognosis and disease management. Recently developed specific monoclonal antibodies (MAbs) targeting cytokines and immune cells are being used for safer treatment and achieving longer remission.
Collapse
Affiliation(s)
- Harinder Singh
- Immunogenomics Laboratory, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| | - Uma Kumar
- Department of Rheumatology, All India Institute of Medical Sciences, New Delhi, India.
| | - Sabyasachi Senapati
- Immunogenomics Laboratory, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| |
Collapse
|
10
|
Chu TY, Zheng-Gérard C, Huang KY, Chang YC, Chen YW, I KY, Lo YL, Chiang NY, Chen HY, Stacey M, Gordon S, Tseng WY, Sun CY, Wu YM, Pan YS, Huang CH, Lin CY, Chen TC, El Omari K, Antonelou M, Henderson SR, Salama A, Seiradake E, Lin HH. GPR97 triggers inflammatory processes in human neutrophils via a macromolecular complex upstream of PAR2 activation. Nat Commun 2022; 13:6385. [PMID: 36302784 PMCID: PMC9613636 DOI: 10.1038/s41467-022-34083-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 10/13/2022] [Indexed: 12/25/2022] Open
Abstract
Neutrophils play essential anti-microbial and inflammatory roles in host defense, however, their activities require tight regulation as dysfunction often leads to detrimental inflammatory and autoimmune diseases. Here we show that the adhesion molecule GPR97 allosterically activates CD177-associated membrane proteinase 3 (mPR3), and in conjugation with several protein interaction partners leads to neutrophil activation in humans. Crystallographic and deletion analysis of the GPR97 extracellular region identified two independent mPR3-binding domains. Mechanistically, the efficient binding and activation of mPR3 by GPR97 requires the macromolecular CD177/GPR97/PAR2/CD16b complex and induces the activation of PAR2, a G protein-coupled receptor known for its function in inflammation. Triggering PAR2 by the upstream complex leads to strong inflammatory activation, prompting anti-microbial activities and endothelial dysfunction. The role of the complex in pathologic inflammation is underscored by the finding that both GPR97 and mPR3 are upregulated on the surface of disease-associated neutrophils. In summary, we identify a PAR2 activation mechanism that directs neutrophil activation, and thus inflammation. The PR3/CD177/GPR97/PAR2/CD16b protein complex, therefore, represents a potential therapeutic target for neutrophil-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Tai-Ying Chu
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | | | - Kuan-Yeh Huang
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chi Chang
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ying-Wen Chen
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kuan-Yu I
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Ling Lo
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Nien-Yi Chiang
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Yi Chen
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Martin Stacey
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Siamon Gordon
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Wen-Yi Tseng
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital-Keelung, Keelung, Taiwan
| | - Chiao-Yin Sun
- Department of Nephrology, Chang Gung Memorial Hospital-Keelung, Keelung, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yen-Mu Wu
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Yi-Shin Pan
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Chien-Hao Huang
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Chun-Yen Lin
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Tse-Ching Chen
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Kamel El Omari
- Diamond Light Source Limited, Harwell Science and Innovation Campus, Didcot, UK
| | | | | | - Alan Salama
- Department of Renal Medicine, Royal Free Campus, UCL, London, UK
| | - Elena Seiradake
- Department of Biochemistry, University of Oxford, Oxford, UK.
| | - Hsi-Hsien Lin
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital-Keelung, Keelung, Taiwan.
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan.
| |
Collapse
|
11
|
Vallejo J, Saigusa R, Gulati R, Armstrong Suthahar SS, Suryawanshi V, Alimadadi A, Durant CP, Ghosheh Y, Roy P, Ehinger E, Pattarabanjird T, Hanna DB, Landay AL, Tracy RP, Lazar JM, Mack WJ, Weber KM, Adimora AA, Hodis HN, Tien PC, Ofotokun I, Heath SL, Shemesh A, McNamara CA, Lanier LL, Hedrick CC, Kaplan RC, Ley K. Combined protein and transcript single-cell RNA sequencing in human peripheral blood mononuclear cells. BMC Biol 2022; 20:193. [PMID: 36045343 PMCID: PMC9434837 DOI: 10.1186/s12915-022-01382-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/01/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Cryopreserved peripheral blood mononuclear cells (PBMCs) are frequently collected and provide disease- and treatment-relevant data in clinical studies. Here, we developed combined protein (40 antibodies) and transcript single-cell (sc)RNA sequencing (scRNA-seq) in PBMCs. RESULTS Among 31 participants in the Women's Interagency HIV Study (WIHS), we sequenced 41,611 cells. Using Boolean gating followed by Seurat UMAPs (tool for visualizing high-dimensional data) and Louvain clustering, we identified 50 subsets among CD4+ T, CD8+ T, B, NK cells, and monocytes. This resolution was superior to flow cytometry, mass cytometry, or scRNA-seq without antibodies. Combined protein and transcript scRNA-seq allowed for the assessment of disease-related changes in transcriptomes and cell type proportions. As a proof-of-concept, we showed such differences between healthy and matched individuals living with HIV with and without cardiovascular disease. CONCLUSIONS In conclusion, combined protein and transcript scRNA sequencing is a suitable and powerful method for clinical investigations using PBMCs.
Collapse
Affiliation(s)
- Jenifer Vallejo
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Ryosuke Saigusa
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Rishab Gulati
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | | | | | - Ahmad Alimadadi
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | | | - Yanal Ghosheh
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Payel Roy
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Erik Ehinger
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Tanyaporn Pattarabanjird
- Carter Immunology Center, Cardiovascular Division, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - David B Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alan L Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Russell P Tracy
- Departments of Pathology & Laboratory Medicine and Biochemistry, University of Vermont Larner College of Medicine, Colchester, VT, USA
| | - Jason M Lazar
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Wendy J Mack
- Department of Medicine and Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Atherosclerosis Research Unit, University of Southern California, Los Angeles, CA, USA
| | - Kathleen M Weber
- Cook County Health/Hektoen Institute of Medicine, Chicago, IL, USA
| | - Adaora A Adimora
- Department of Medicine, University of North Carolina School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Howard N Hodis
- Department of Medicine and Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Atherosclerosis Research Unit, University of Southern California, Los Angeles, CA, USA
| | - Phyllis C Tien
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Igho Ofotokun
- Department of Medicine, Infectious Disease Division and Grady Health Care System, Emory University School of Medicine, Atlanta, GA, USA
| | - Sonya L Heath
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Avishai Shemesh
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Coleen A McNamara
- Carter Immunology Center, Cardiovascular Division, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Lewis L Lanier
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Catherine C Hedrick
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Fred Hutchinson Cancer Research Center, Public Health Sciences Division, Seattle, WA, USA
| | - Klaus Ley
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA.
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA.
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
12
|
Mohamed MS, Ghaly S, Azmy KH, Mohamed GA. Assessment of interleukin 32 as a novel biomarker for non-alcoholic fatty liver disease. EGYPTIAN LIVER JOURNAL 2022. [DOI: 10.1186/s43066-022-00189-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Non-alcoholic fatty liver disease (NAFLD) is a metabolic disorder characterised by enhanced hepatic fat deposition and inflammation. Efforts to manage NAFLD are limited by the poorly characterised pathological processes and the lack of precise non-invasive markers, thus, proving the need to further study the involved cytokines, which, in turn, may represent novel molecular targets with possible diagnostic and therapeutic applications. Hence, we aimed to assess the diagnostic utility of serum interleukin 32 (IL-32) in NAFLD cases. This case-control study included 40 NAFLD patients and 40 healthy controls. The serum IL-32 concentrations were assessed by the enzyme-linked immunosorbent assay (ELISA).
Results
The serum IL-32 concentrations were significantly higher in NAFLD cases than controls (76 [45.5–111.125] vs. 13 [8–15] pg/mL, P < 0.001, respectively). IL-32 at a cut-off point > 22.5 pg/mL had 100% sensitivity, 87.50% specificity, 88.9% positive predictive value, 100% negative predictive value, and 98.2% accuracy in detecting the NAFLD cases.
Conclusion
Serum IL-32 could be considered a novel non-invasive marker for NAFLD. Further investigations are warranted to verify the potential utility of IL-32 in the clinical setting.
Collapse
|
13
|
Liu Y, Yan H, Gu H, Zhang E, He J, Cao W, Qu J, Xu R, Cao L, He D, Zhang J, Hou Y, Cai Z. Myeloma-derived IL-32γ induced PD-L1 expression in macrophages facilitates immune escape via the PFKFB3-JAK1 axis. Oncoimmunology 2022; 11:2057837. [PMID: 35371618 PMCID: PMC8973380 DOI: 10.1080/2162402x.2022.2057837] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Yang Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Haimeng Yan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Huiyao Gu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Enfan Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Jingsong He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Wen Cao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Jianwei Qu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Ruyi Xu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Liqin Cao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Donghua He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Jinna Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Yifan Hou
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Ribeiro-Dias F, Oliveira I. A Critical Overview of Interleukin 32 in Leishmaniases. Front Immunol 2022; 13:849340. [PMID: 35309341 PMCID: PMC8927017 DOI: 10.3389/fimmu.2022.849340] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/11/2022] [Indexed: 12/22/2022] Open
Abstract
Interleukin-32 (IL-32) has several immune regulatory properties, which have driven its investigation in the context of various diseases. IL-32 expression is reported to be induced in the lesions of patients with American tegumentary leishmaniasis (ATL) by the New World Leishmania spp. that are responsible for causing ATL and visceral leishmaniasis (VL). IL-32 expression may elevate the inflammatory process through the induction of pro-inflammatory cytokines and also via mechanisms directed to kill the parasites. The genetic variants of IL-32 might be associated with the resistance or susceptibility to ATL, while different isoforms of IL-32 could be associated with distinct T helper lymphocyte profiles. IL-32 also determines the transcriptional profile in the bone marrow progenitor cells to mediate the trained immunity induced by β-glucan and BCG, thereby contributing to the resistance against Leishmania. IL-32γ is essential for the vitamin D-dependent microbicidal pathway for parasite control. In this context, the present review report briefly discusses the data retrieved from the studies conducted on IL-32 in leishmaniasis in humans and mice to highlight the current challenges to understanding the role of IL-32 in leishmaniasis.
Collapse
Affiliation(s)
- Fátima Ribeiro-Dias
- Laboratório de Imunidade Natural, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | | |
Collapse
|
15
|
Calcium Dobesilate Modulates PKCδ-NADPH Oxidase- MAPK-NF-κB Signaling Pathway to Reduce CD14, TLR4, and MMP9 Expression during Monocyte-to-Macrophage Differentiation: Potential Therapeutic Implications for Atherosclerosis. Antioxidants (Basel) 2021; 10:antiox10111798. [PMID: 34829669 PMCID: PMC8615002 DOI: 10.3390/antiox10111798] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 01/18/2023] Open
Abstract
Monocyte-to-macrophage differentiation results in the secretion of various inflammatory mediators and oxidative stress molecules necessary for atherosclerosis pathogenesis. Consequently, this differentiation represents a potential clinical target in atherosclerosis. Calcium dobesilate (CaD), an established vasoactive and angioprotective drug in experimental models of diabetic microvascular complications reduces oxidative stress and inhibits inflammation via diverse molecular targets; however, its effect on monocytes/macrophages is poorly understood. In this study, we investigated the anti-inflammatory mechanism of CaD during phorbol 12-myristate 13-acetate (PMA)-induced monocyte-to-macrophage differentiation in in vitro models of sepsis (LPS) and hyperglycemia, using THP-1 monocytic cell line. CaD significantly suppressed CD14, TLR4, and MMP9 expression and activity, lowering pro-inflammatory mediators, such as IL1β, TNFα, and MCP-1. The effects of CaD translated through to studies on primary human macrophages. CaD inhibited reactive oxygen species (ROS) generation, PKCδ, MAPK (ERK1/2 and p38) phosphorylation, NOX2/p47phox expression, and membrane translocation. We used hydrogen peroxide (H2O2) to mimic oxidative stress, demonstrating that CaD suppressed PKCδ activation via its ROS-scavenging properties. Taken together, we demonstrate for the first time that CaD suppresses CD14, TLR4, MMP9, and signature pro-inflammatory cytokines, in human macrophages, via the downregulation of PKCδ/NADPH oxidase/ROS/MAPK/NF-κB-dependent signaling pathways. Our data present novel mechanisms of how CaD alleviates metabolic and infectious inflammation.
Collapse
|
16
|
Liu YJ, Li JP, Zhang Y, Nie MJ, Zhang YH, Liu SL, Zou X. FSTL3 is a Prognostic Biomarker in Gastric Cancer and is Correlated with M2 Macrophage Infiltration. Onco Targets Ther 2021; 14:4099-4117. [PMID: 34262295 PMCID: PMC8274543 DOI: 10.2147/ott.s314561] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/22/2021] [Indexed: 12/31/2022] Open
Abstract
Purpose Follistatin-related gene 3 (FSTL3), an established oncogene, can modulate target gene expression via members of the transforming growth factor β (TGF-β) superfamily. The present study was conducted to evaluate the expression of FSTL3 in gastric cancer (GC) and to determine its prognostic significance. We also evaluated the possible mechanisms involved in the oncogenic role of FSTL3 in gastric carcinogenesis and development. Methods We obtained data from the Human Protein Atlas, MethSurv, cBioPortal, UALCAN, TIMER, GEPIA, STRING, GeneMANIA, ONCOMINE, and MEXPRESS databases and examined it using R software. RNAi was used to establish stable FSTL3-knockdown (shFSTL3) and overexpression (OE) cell strains. Western blot; enzyme-linked immunosorbent (ELISA); and immunohistochemical (ICH), immunofluorescence, and phalloidin staining were used for examining protein expression. Cell invasion and migration were determined using transwell and scratch-wound assays. After tumor-associated macrophage (TAM) generation, co-culturing of cancer cells with TAMs was performed to confirm the relationship between FSTL3 and TAMs. Results In GC patients, FSTL3 mRNA and protein levels were upregulated. FSTL3 expression was significantly linked to cancer stage as well as to pathological tumor grade in GC. Moreover, a high expression of FSTL3 was associated with a dismal survival duration in patients with GC. Furthermore, functional enrichment analysis demonstrated that FSTL3 overexpression could activate epithelial-mesenchymal transition (EMT) by promoting F-actin expression and BMP/SMAD signaling. Finally, immunofluorescence staining confirmed that the overexpression of FSTL3 promoted the proliferation of M2 TAMs. Conclusion Taken together, our findings suggest that FSTL3 may be involved in GC progression via the promotion of BMP/SMAD signaling-mediated EMT and M2 macrophage activation.
Collapse
Affiliation(s)
- Yuan-Jie Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Jie-Pin Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China.,Department of Oncology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, 215600, People's Republic of China
| | - Ying Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Meng-Jun Nie
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Yong-Hua Zhang
- Department of Oncology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, 215600, People's Republic of China
| | - Shen-Lin Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Xi Zou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China
| |
Collapse
|
17
|
Bunet R, Nayrac M, Ramani H, Sylla M, Durand M, Chartrand-Lefebvre C, Routy JP, Landay AL, Gauchat JF, Chomont N, Ancuta P, Kaufmann DE, Bernard N, Tremblay CL, El-Far M. Loss of CD96 Expression as a Marker of HIV-Specific CD8 + T-Cell Differentiation and Dysfunction. Front Immunol 2021; 12:673061. [PMID: 34122431 PMCID: PMC8190400 DOI: 10.3389/fimmu.2021.673061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/11/2021] [Indexed: 01/01/2023] Open
Abstract
Persistent immune activation and inflammation in people living with HIV (PLWH) are associated with immunosenescence, premature aging and increased risk of non-AIDS comorbidities, with the underlying mechanisms not fully understood. In this study, we show that downregulation of the T-cell immunoglobulin receptor CD96 on CD8+ T cells from PLWH is associated with decreased expression of the co-stimulatory receptors CD27 and CD28, higher expression of the senescence marker CD57 and accumulation of a terminally differentiated T-cell memory phenotype. In addition, we show that CD96-low CD8+ T-cells display lower proliferative potential compared to their CD96-high counterparts and that loss of CD96 expression by HIV-specific CD8+ T-cells is associated with a suboptimal response to HIV antigens. In conclusion, our results suggest that CD96 marks CD8+ T-cells with competent responses to HIV and the loss of its expression might be used as a biomarker for CD8+ T-cell senescence and dysfunction in PLWH.
Collapse
Affiliation(s)
- Rémi Bunet
- CHUM-Research Centre, Montréal, Montréal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Manon Nayrac
- CHUM-Research Centre, Montréal, Montréal, QC, Canada
| | - Hardik Ramani
- CHUM-Research Centre, Montréal, Montréal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Mohamed Sylla
- CHUM-Research Centre, Montréal, Montréal, QC, Canada
| | - Madeleine Durand
- CHUM-Research Centre, Montréal, Montréal, QC, Canada.,Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | | | - Jean-Pierre Routy
- Research Institute of McGill University Health Centre, Montréal, QC, Canada
| | - Alan L Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Jean-Francois Gauchat
- Faculté de Médecine, Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada
| | - Nicolas Chomont
- CHUM-Research Centre, Montréal, Montréal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Petronela Ancuta
- CHUM-Research Centre, Montréal, Montréal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Daniel E Kaufmann
- CHUM-Research Centre, Montréal, Montréal, QC, Canada.,Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Nicole Bernard
- Research Institute of McGill University Health Centre, Montréal, QC, Canada
| | - Cécile L Tremblay
- CHUM-Research Centre, Montréal, Montréal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | | |
Collapse
|
18
|
IL32: The multifaceted and unconventional cytokine. Hum Immunol 2021; 82:659-667. [PMID: 34024634 DOI: 10.1016/j.humimm.2021.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/20/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
Interleukin 32 is a unique intracellular cytokine which affects many cellular and physiological functions like cell death and survival, inflammation and response to pathogens. With numerous transcripts, more than one biologically active isoforms, IL32 drives its effect in diverse cellular functions. A cytokine restricted to higher mammals, it is known to fine tune multiple pathways involved in metabolic processes or infection. It modulates the immune response against diverse pathogens like Leishmania, Mycobacterium and HIV. IL32 has been associated with cancers of inflammatory nature too. It also plays an important role in chronic inflammatory diseases like RA, lung and airway disease like COPD. In this review we have discussed about identification and characterization of this non classical cytokine IL32, its structure and function at gene as well as at protein level, isoforms and their diverse functions. Role of IL32 in multiple diseases and particularly mycobacterial disease has been highlighted here. We have also summarised the genetic variants present in the IL32 gene and it's promoter region. Association of these variants, with cellular phenotype, patho-physiological conditions in different disease have also been discussed here.
Collapse
|
19
|
Bianco C, Casirati E, Malvestiti F, Valenti L. Genetic predisposition similarities between NASH and ASH: Identification of new therapeutic targets. JHEP Rep 2021; 3:100284. [PMID: 34027340 PMCID: PMC8122117 DOI: 10.1016/j.jhepr.2021.100284] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Fatty liver disease can be triggered by a combination of excess alcohol, dysmetabolism and other environmental cues, which can lead to steatohepatitis and can evolve to acute/chronic liver failure and hepatocellular carcinoma, especially in the presence of shared inherited determinants. The recent identification of the genetic causes of steatohepatitis is revealing new avenues for more effective risk stratification. Discovery of the mechanisms underpinning the detrimental effect of causal mutations has led to some breakthroughs in the comprehension of the pathophysiology of steatohepatitis. Thanks to this approach, hepatocellular fat accumulation, altered lipid droplet remodelling and lipotoxicity have now taken centre stage, while the role of adiposity and gut-liver axis alterations have been independently validated. This process could ignite a virtuous research cycle that, starting from human genomics, through omics approaches, molecular genetics and disease models, may lead to the development of new therapeutics targeted to patients at higher risk. Herein, we also review how this knowledge has been applied to: a) the study of the main PNPLA3 I148M risk variant, up to the stage of the first in-human therapeutic trials; b) highlight a role of MBOAT7 downregulation and lysophosphatidyl-inositol in steatohepatitis; c) identify IL-32 as a candidate mediator linking lipotoxicity to inflammation and liver disease. Although this precision medicine drug discovery pipeline is mainly being applied to non-alcoholic steatohepatitis, there is hope that successful products could be repurposed to treat alcohol-related liver disease as well.
Collapse
Key Words
- AA, arachidonic acid
- ASH, alcoholic steatohepatitis
- DAG, diacylglycerol
- DNL, de novo lipogenesis
- ER, endoplasmic reticulum
- FFAs, free fatty acids
- FGF19, fibroblast growth factor 19
- FLD, fatty liver disease
- FXR, farnesoid X receptor
- GCKR, glucokinase regulator
- GPR55, G protein-coupled receptor 55
- HCC, hepatocellular carcinoma
- HFE, homeostatic iron regulator
- HSC, hepatic stellate cells
- HSD17B13, hydroxysteroid 17-beta dehydrogenase 13
- IL-, interleukin-
- IL32
- LDs, lipid droplets
- LPI, lysophosphatidyl-inositol
- MARC1, mitochondrial amidoxime reducing component 1
- MBOAT7
- MBOAT7, membrane bound O-acyltransferase domain-containing 7
- NASH, non-alcoholic steatohepatitis
- PNPLA3
- PNPLA3, patatin like phospholipase domain containing 3
- PPAR, peroxisome proliferator-activated receptor
- PRS, polygenic risk score
- PUFAs, polyunsaturated fatty acids
- SREBP, sterol response element binding protein
- TAG, triacylglycerol
- TNF-α, tumour necrosis factor-α
- alcoholic liver disease
- cirrhosis
- fatty liver disease
- genetics
- interleukin-32
- non-alcoholic fatty liver disease
- precision medicine
- steatohepatitis
- therapy
Collapse
Affiliation(s)
- Cristiana Bianco
- Precision Medicine - Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elia Casirati
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Francesco Malvestiti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Luca Valenti
- Precision Medicine - Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
20
|
Baselli GA, Dongiovanni P, Rametta R, Meroni M, Pelusi S, Maggioni M, Badiali S, Pingitore P, Maurotti S, Montalcini T, Taliento AE, Prati D, Rossi G, Fracanzani AL, Mancina RM, Romeo S, Valenti L. Liver transcriptomics highlights interleukin-32 as novel NAFLD-related cytokine and candidate biomarker. Gut 2020; 69:1855-1866. [PMID: 32001554 PMCID: PMC7497582 DOI: 10.1136/gutjnl-2019-319226] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 12/05/2019] [Accepted: 12/22/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Efforts to manage non-alcoholic fatty liver disease (NAFLD) are limited by the incomplete understanding of the pathogenic mechanisms and the absence of accurate non-invasive biomarkers. The aim of this study was to identify novel NAFLD therapeutic targets andbiomarkers by conducting liver transcriptomic analysis in patients stratified by the presence of the PNPLA3 I148M genetic risk variant. DESIGN We sequenced the hepatic transcriptome of 125 obese individuals. 'Severe NAFLD' was defined as the presence of steatohepatitis, NAFLD activity score ≥4 or fibrosis stage ≥2. The circulating levels of the most upregulated transcript, interleukin-32 (IL32), were measured by ELISA. RESULTS Carriage of the PNPLA3 I148M variant correlated with the two major components of hepatic transcriptome variability and broadly influenced gene expression. In patients with severe NAFLD, there was an upregulation of inflammatory and lipid metabolism pathways. IL32 was the most robustly upregulated gene in the severe NAFLD group (adjusted p=1×10-6), and its expression correlated with steatosis severity, both in I148M variant carriers and non-carriers. In 77 severely obese, and in a replication cohort of 160 individuals evaluated at the hepatology service, circulating IL32 levels were associated with both NAFLD and severe NAFLD independently of aminotransferases (p<0.01 for both). A linear combination of IL32-ALT-AST showed a better performance than ALT-AST alone in NAFLD diagnosis (area under the curve=0.92 vs 0.81, p=5×10-5). CONCLUSION Hepatic IL32 is overexpressed in NAFLD, correlates with hepatic fat and liver damage, and is detectable in the circulation, where it is independently associated with the presence and severity of NAFLD.
Collapse
Affiliation(s)
- Guido Alessandro Baselli
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milano, Lombardia, Italy,Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| | - Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| | - Raffaela Rametta
- General Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| | - Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| | - Serena Pelusi
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milano, Lombardia, Italy,Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| | - Marco Maggioni
- Pathology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| | - Sara Badiali
- Surgery, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| | - Piero Pingitore
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Calabria, Italy
| | - Samantha Maurotti
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Calabria, Italy
| | - Tiziana Montalcini
- Department of Clinical and Experimental Medicine, Nutrition Unit, Magna Graecia University of Catanzaro, Catanzaro, Calabria, Italy
| | - Alice Emma Taliento
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| | - Daniele Prati
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| | - Giorgio Rossi
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milano, Lombardia, Italy,Liver Transplantation Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Anna Ludovica Fracanzani
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milano, Lombardia, Italy,General Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| | | | - Stefano Romeo
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Calabria, Italy .,Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory, Cardiology Department, University of Gothenburg, Goteborg, Sweden
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milano, Lombardia, Italy .,Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| |
Collapse
|
21
|
Rayees S, Rochford I, Joshi JC, Joshi B, Banerjee S, Mehta D. Macrophage TLR4 and PAR2 Signaling: Role in Regulating Vascular Inflammatory Injury and Repair. Front Immunol 2020; 11:2091. [PMID: 33072072 PMCID: PMC7530636 DOI: 10.3389/fimmu.2020.02091] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages play a central role in dictating the tissue response to infection and orchestrating subsequent repair of the damage. In this context, macrophages residing in the lungs continuously sense and discriminate among a wide range of insults to initiate the immune responses important to host-defense. Inflammatory tissue injury also leads to activation of proteases, and thereby the coagulation pathway, to optimize injury and repair post-infection. However, long-lasting inflammatory triggers from macrophages can impair the lung's ability to recover from severe injury, leading to increased lung vascular permeability and neutrophilic injury, hallmarks of Acute Lung Injury (ALI). In this review, we discuss the roles of toll-like receptor 4 (TLR4) and protease activating receptor 2 (PAR2) expressed on the macrophage cell-surface in regulating lung vascular inflammatory signaling.
Collapse
Affiliation(s)
- Sheikh Rayees
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, United States
| | - Ian Rochford
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, United States
| | - Jagdish Chandra Joshi
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, United States
| | - Bhagwati Joshi
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, United States
| | - Somenath Banerjee
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, United States
| | - Dolly Mehta
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, United States
| |
Collapse
|
22
|
Aass KR, Kastnes MH, Standal T. Molecular interactions and functions of IL-32. J Leukoc Biol 2020; 109:143-159. [PMID: 32869391 DOI: 10.1002/jlb.3mr0620-550r] [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] [Received: 03/02/2020] [Revised: 06/29/2020] [Accepted: 07/28/2020] [Indexed: 12/16/2022] Open
Abstract
IL-32 is a multifaceted cytokine associated with several diseases and inflammatory conditions. Its expression is induced in response to cellular stress such as hypoxia, infections, and pro-inflammatory cytokines. IL-32 can be secreted from cells and can induce the production of pro-inflammatory cytokines from several cell types but are also described to have anti-inflammatory functions. The intracellular form of IL-32 is shown to play an important role in various cellular processes, including the defense against intracellular bacteria and viruses and in modulation of cell metabolism. In this review, we discuss current literature on molecular interactions of IL-32 with other proteins. We also review data on the role of intracellular IL-32 as a metabolic regulator and its role in antimicrobial host defense.
Collapse
Affiliation(s)
- Kristin Roseth Aass
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research (CEMIR), Trondheim, Norway
| | - Martin H Kastnes
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research (CEMIR), Trondheim, Norway
| | - Therese Standal
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research (CEMIR), Trondheim, Norway.,Department of Hematology, St. Olavs Hospital, Trondheim, Norway
| |
Collapse
|
23
|
Wu X, Lee B, Zhu L, Ding Z, Chen Y. Exposure to mold proteases stimulates mucin production in airway epithelial cells through Ras/Raf1/ERK signal pathway. PLoS One 2020; 15:e0231990. [PMID: 32320453 PMCID: PMC7176129 DOI: 10.1371/journal.pone.0231990] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 04/03/2020] [Indexed: 10/25/2022] Open
Abstract
Environmental mold (fungus) exposure poses a significant threat to public health by causing illnesses ranging from invasive fungal diseases in immune compromised individuals to allergic hypertensive diseases such as asthma and asthma exacerbation in otherwise healthy people. However, the molecular pathogenesis has not been completely understood, and treatment options are limited. Due to its thermo-tolerance to the normal human body temperature, Aspergillus. fumigatus (A.fumigatus) is one of the most important human pathogens to cause different lung fungal diseases including fungal asthma. Airway obstruction and hyperresponsiveness caused by mucus overproduction are the hallmarks of many A.fumigatus induced lung diseases. To understand the underlying molecular mechanism, we have utilized a well-established A.fumigatus extracts (AFE) model to elucidate downstream signal pathways that mediate A.fumigatus induced mucin production in airway epithelial cells. AFE was found to stimulate time- and dose-dependent increase of major airway mucin gene expression (MUC5AC and MUC5B) partly via the elevation of their promoter activities. We also demonstrated that EGFR was required but not sufficient for AFE-induced mucin expression, filling the paradoxical gap from a previous study using the same model. Furthermore, we showed that fungal proteases in AFE were responsible for mucin induction by activating a Ras/Raf1/ERK signaling pathway. Ca2+ signaling, but ROS, both of which were stimulated by fungal proteases, was an indispensable determinant for ERK activation and mucin induction. The discovery of this novel pathway likely contributes to our understanding of the pathogenesis of fungal sensitization in allergic diseases such as fungal asthma.
Collapse
Affiliation(s)
- Xianxian Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona, United States of America
| | - Boram Lee
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona, United States of America
| | - Lingxiang Zhu
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona, United States of America
| | - Zhi Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
- Changzhou High-Tech Research Institute of Nanjing University, Changzhou, China
- * E-mail: (ZD); (YC)
| | - Yin Chen
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona, United States of America
- Asthma & Airway Disease Research Center, University of Arizona, Tucson, Arizona, United States of America
- * E-mail: (ZD); (YC)
| |
Collapse
|
24
|
Pavlovic M, Jovanovic I, Arsenijevic N. Interleukin-32 in Infection, Inflammation and Cancer Biology. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2020. [DOI: 10.1515/sjecr-2016-0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Cytokines are small pleiotropic polypeptids secreted dominantly by the cells of the immune system. These polypeptids are main mediators of innate and acquired immunity, responsible for clonal expansion and differentiation of immune cells, initiation of immune response and enhancing of effector functions of leukocytes. Cytokine-related effects are most studied in the fields of inflammation, immunology, and cancer biology. In this review we discuss one of the most intriguing, recently discovered proinflammatory cytokine, interleukin 32.
Collapse
Affiliation(s)
- Mladen Pavlovic
- Department of Surgery, Faculty of Medical Sciences , University of Kragujevac , Serbia
| | - Ivan Jovanovic
- Center for Molecular Medicine and Stem Cell Research , Faculty of Medical Sciences , University of Kragujevac , Serbia
| | - Nebojsa Arsenijevic
- Center for Molecular Medicine and Stem Cell Research , Faculty of Medical Sciences , University of Kragujevac , Serbia
| |
Collapse
|
25
|
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: 15] [Impact Index Per Article: 2.5] [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
|
26
|
Yan H, Dong M, Liu X, Shen Q, He D, Huang X, Zhang E, Lin X, Chen Q, Guo X, Chen J, Zheng G, Wang G, He J, Yi Q, Cai Z. Multiple myeloma cell-derived IL-32γ increases the immunosuppressive function of macrophages by promoting indoleamine 2,3-dioxygenase (IDO) expression. Cancer Lett 2019; 446:38-48. [PMID: 30660652 DOI: 10.1016/j.canlet.2019.01.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 12/14/2018] [Accepted: 01/08/2019] [Indexed: 12/28/2022]
Abstract
The interaction of multiple myeloma (MM) cells with macrophages (MΦs) contributes to the pathophysiology of MM. We previously showed that IL-32 is overexpressed in MM patients. The present study was designed to explore the clinical significance of IL-32 in MM and to further elucidate the mechanisms underlying the IL-32-mediated immune function of MΦs. Our results showed that high IL-32 expression in MM patients was associated with more advanced clinical stage. RNA-sequencing revealed that IL-32γ significantly induced the production of the immunosuppressive molecule indoleamine 2,3-dioxygenase (IDO) in MΦs, and this effect was verified by qRT-PCR, western blotting, and immunofluorescence. Furthermore, MM cells with IL-32-knockdown showed a reduced ability to promote IDO expression. As a binding protein for IL-32, proteinase 3 (PR3) was universally expressed on the surfaces of MΦs, and knockdown of PR3 or inhibition of the STAT3 and NF-κB pathways hindered the IL-32γ-mediated stimulation of IDO expression. Finally, IDO-positive IL-32γ-educated MΦs inhibited CD4+ T cell proliferation and IL-2, IFN-γ, and TNF-α production. Taken together, our results indicate that IL-32γ derived from MM cells promotes the immunosuppressive function of MΦs and is a potential target for MM treatment.
Collapse
Affiliation(s)
- Haimeng Yan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mengmeng Dong
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinling Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiang Shen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Donghua He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xi Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Enfan Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xuanru Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qingxiao Chen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xing Guo
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jing Chen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Gaofeng Zheng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Gang Wang
- Department of Hematology, People's Hospital of Quzhou, Quzhou, Zhejiang, China
| | - Jingsong He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qing Yi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Zhen Cai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Institute of Hematology, Zhejiang University, China.
| |
Collapse
|
27
|
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: 0.6] [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]
|
28
|
The Biology and Role of Interleukin-32 in Tuberculosis. J Immunol Res 2018; 2018:1535194. [PMID: 30426023 PMCID: PMC6217754 DOI: 10.1155/2018/1535194] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/08/2018] [Accepted: 08/29/2018] [Indexed: 12/31/2022] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis, remains a leading cause of morbidity and mortality globally, with nearly 10.4 million new cases of incidence and over 1.7 million deaths annually. Drug-resistant M. tuberculosis strains, especially multidrug-resistant or extensively drug-resistant strains, have further intensified the problem associated with tuberculosis control. Host-directed therapy is a promising alternative for tuberculosis control. IL-32 is increasingly recognized as an important host molecule against tuberculosis. In this review, we highlight the proinflammatory properties of IL-32 and the mode of action of IL-32 in mycobacterial infections to inspire the development of novel immunity-based countermeasures and host-directed therapies against tuberculosis.
Collapse
|
29
|
Dos Santos JC, Damen MSMA, Joosten LAB, Ribeiro-Dias F. Interleukin-32: An endogenous danger signal or master regulator of intracellular pathogen infections-Focus on leishmaniases. Semin Immunol 2018; 38:15-23. [PMID: 29551246 DOI: 10.1016/j.smim.2018.02.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 02/23/2018] [Indexed: 02/07/2023]
Abstract
Interleukin 32 (IL-32) is an intracellular cytokine produced by immune and non immune cells after different stimuli. It contributes to inflammation and control of intracellular pathogens mainly by inducing proinflammatory cytokines and microbicidal molecules. Evidence is rising showing that IL-32 can be considered an endogenous danger signal after tissue injury, amplifying the inflammatory process and acquired immune responses. It seems to be a master regulator of intracellular infectious diseases. In this review, first the general properties of IL-32 are described followed by its role in the immunopathogenesis of inflammatory and infectious diseases. Roles of IL-32 in the control of infectious diseases caused by intracellular pathogens are reported, and later a focus on IL-32 in leishmaniases, diseases caused by an intracellular protozoan, is presented.
Collapse
Affiliation(s)
- Jéssica C Dos Santos
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands; Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Michelle S M A Damen
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands; Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Fátima Ribeiro-Dias
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil.
| |
Collapse
|
30
|
|
31
|
Chávez-Galán L, Ramon-Luing L, Carranza C, Garcia I, Sada-Ovalle I. Lipoarabinomannan Decreases Galectin-9 Expression and Tumor Necrosis Factor Pathway in Macrophages Favoring Mycobacterium tuberculosis Intracellular Growth. Front Immunol 2017; 8:1659. [PMID: 29230224 PMCID: PMC5711832 DOI: 10.3389/fimmu.2017.01659] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 11/13/2017] [Indexed: 12/24/2022] Open
Abstract
Lipoarabinomannan (LAM) is a lipid virulent factor secreted by Mycobacterium tuberculosis (Mtb). LAM can be found in the sputum and urine of patients with active tuberculosis. When human monocytes are differentiated into macrophages [monocyte-derived macrophages (MDM)] in the presence of LAM, MDM are poorly functional which may limit the immune response to Mtb infection. Our previous studies have shown that TIM3 and galectin (GAL)9 interaction induces anti-mycobacterial activity, and the expression levels of TIM3 and GAL9 are downregulated during Mtb infection. We postulated that LAM affects GAL9/TIM3 pathway, and, in consequence, the ability of the macrophage to control bacterial growth could be affected. In this work, we have generated MDM in the presence of LAM and observed that the expression of TIM3 was not affected; in contrast, GAL9 expression was downregulated at the transcriptional and protein levels. We observed that the cell surface and the soluble form of tumor necrosis factor (TNF) receptor 2 were decreased. We also found that when LAM-exposed MDM were activated with LPS, they produced less TNF, and the transcription factor proteinase-activated receptor-2 (PAR2), which is involved in host immune responses to infection, was not induced. Our data show that LAM-exposed MDM were deficient in the control of intracellular growth of Mtb. In conclusion, LAM-exposed MDM leads to MDM with impaired intracellular signal activation affecting GAL9, TNF, and PAR2 pathways, which are important to restrict Mtb growth.
Collapse
Affiliation(s)
- Leslie Chávez-Galán
- Laboratory of Integrative Immunology, National Institute of Respiratory Diseases Ismael Cosio Villegas, Mexico City, Mexico
| | - Lucero Ramon-Luing
- Laboratory of Integrative Immunology, National Institute of Respiratory Diseases Ismael Cosio Villegas, Mexico City, Mexico
| | - Claudia Carranza
- Department of Microbiology, National Institute of Respiratory Diseases Ismael Cosio Villegas, Mexico City, Mexico
| | - Irene Garcia
- Department of Pathology and Immunology, Centre Medical Universitaire, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Isabel Sada-Ovalle
- Laboratory of Integrative Immunology, National Institute of Respiratory Diseases Ismael Cosio Villegas, Mexico City, Mexico
| |
Collapse
|
32
|
Interleukin-32α promotes the proliferation of multiple myeloma cells by inducing production of IL-6 in bone marrow stromal cells. Oncotarget 2017; 8:92841-92854. [PMID: 29190960 PMCID: PMC5696226 DOI: 10.18632/oncotarget.21611] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 08/17/2017] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is a malignant plasma disease closely associated with inflammation. In MM bone marrow microenvironment, bone marrow stromal cells (BMSCs) are the primary source of interleukin-6 (IL-6) secretion, which promotes the proliferation and progression of MM cells. However, it is still unknown how the microenvironment stimulates BMSCs to secrete IL-6. Interleukin-32 (IL-32) is a newly identified pro-inflammatory factor. It was reported that in solid tumors, IL-32 induces changes in other inflammatory factors including IL-6, IL-10, and TNF-α. The aim of this study was to investigate the expression of IL-32 and the role of IL-32 in the MM bone marrow microenvironment. Our data illustrate that MM patients have higher expression of IL-32 than healthy individuals in both bone marrow and peripheral blood. We used ELISA and qRT-PCR to find that malignant plasma cells are the primary source of IL-32 production in MM bone marrow. ELISA and Western blot analysis revealed that recombinant IL-32α induces production of IL-6 in BMSCs by activating NF-κB and STAT3 signaling pathways, konckdown of IL-32 receptor PR3 inhibit this process. Knockdown of IL-32 by shRNA decreased the proliferation in MM cells that induced by BMSCs. In conclusion, IL-32 secreted from MM cells has paracrine effect to induce production of IL-6 in BMSCs, thus feedback to promote MM cells growth.
Collapse
|
33
|
Gorvel L, Korenfeld D, Tung T, Klechevsky E. Dendritic Cell-Derived IL-32α: A Novel Inhibitory Cytokine of NK Cell Function. THE JOURNAL OF IMMUNOLOGY 2017; 199:1290-1300. [PMID: 28701509 DOI: 10.4049/jimmunol.1601477] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 06/11/2017] [Indexed: 12/15/2022]
Abstract
Cytokines produced by dendritic cells (DCs) can largely determine the direction of immunity. Transcriptional analysis revealed that besides IL-15, IL-32 was the only other cytokine expressed by human Langerhans cells. IL-32 is a human cytokine that exists in four main isoforms. Currently, little is known about the regulation and function of the various IL-32 isoforms. In this study, we found that IL-15 is a potent inducer of IL-32α in DCs. Because IL-15 promotes NK cell activation, we investigated the interplay between IL-32 and IL-15 and their role in NK cell activity. We show that IL-32α acts on NK cells to inhibit IL-15-mediated STAT5 phosphorylation and to suppress their IL-15-induced effector molecule expression and cytolytic capacity. IL-32α also acted on DCs by downregulating IL-15-induced IL-18 production, an important cytokine in NK cell activity. Blocking IL-32α during DC:NK cell coculture enhanced NK cell effector molecule expression as well as their cytolytic capacity. Taken together, our findings suggest a feedback inhibition of IL-15-mediated NK cell activity by IL-32α.
Collapse
Affiliation(s)
- Laurent Gorvel
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Daniel Korenfeld
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Thomas Tung
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110
| | - Eynav Klechevsky
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
| |
Collapse
|
34
|
dos Santos JC, Heinhuis B, Gomes RS, Damen MSMA, Real F, Mortara RA, Keating ST, Dinarello CA, Joosten LAB, Ribeiro-Dias F. Cytokines and microbicidal molecules regulated by IL-32 in THP-1-derived human macrophages infected with New World Leishmania species. PLoS Negl Trop Dis 2017; 11:e0005413. [PMID: 28241012 PMCID: PMC5344527 DOI: 10.1371/journal.pntd.0005413] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 03/09/2017] [Accepted: 02/15/2017] [Indexed: 01/08/2023] Open
Abstract
Background Interleukin-32 (IL-32) is expressed in lesions of patients with American Tegumentary Leishmaniasis (ATL), but its precise role in the disease remains unknown. Methodology/Principal findings In the present study, silencing and overexpression of IL-32 was performed in THP-1-derived macrophages infected with Leishmania (Viannia) braziliensis or L. (Leishmania) amazonensis to investigate the role of IL-32 in infection. We report that Leishmania species induces IL-32γ, and show that intracellular IL-32γ protein production is dependent on endogenous TNFα. Silencing or overexpression of IL-32 demonstrated that this cytokine is closely related to TNFα and IL-8. Remarkably, the infection index was augmented in the absence of IL-32 and decreased in cells overexpressing this cytokine. Mechanistically, these effects can be explained by nitric oxide cathelicidin and β-defensin 2 production regulated by IL-32. Conclusions Thus, endogenous IL-32 is a crucial cytokine involved in the host defense against Leishmania parasites. Leishmania (V.) braziliensis and L. (L.) amazonensis are protozoa that infect macrophages and cause cutaneous and mucosal leishmaniasis. Here we showed that both Leishmania species induce the production of IL-32γ in human macrophages. This intracellular and pro-inflammatory cytokine mediates the production of cytokines, especially TNFα and IL-8, in Leishmania-infected macrophages. Differential effects of IL-32γ on TNFα, IL-10 and IL-1Ra production after infection with distinct Leishmania species were detected, consistent with the concept that IL-32γ can differently influence the outcome of inflammatory process in leishmaniasis lesions. Moreover, IL-32γ upregulates microbicidal molecules, antimicrobial peptides, as well as NO, which are known as important factors in parasite control. These results underscore IL-32γ as a crucial cytokine to host defense against leishmaniasis.
Collapse
Affiliation(s)
- Jéssica Cristina dos Santos
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Bas Heinhuis
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rodrigo Saar Gomes
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Michelle S. M. A. Damen
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fernando Real
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Brazil
| | - Renato A. Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Brazil
| | - Samuel T. Keating
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Charles A. Dinarello
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
- School of Medicine, Division of infectious diseases, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Leo A. B. Joosten
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fátima Ribeiro-Dias
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
- * E-mail:
| |
Collapse
|
35
|
Proteinase-activated receptor 2 is involved in the behavioural changes associated with sickness behaviour. J Neuroimmunol 2016; 295-296:139-47. [DOI: 10.1016/j.jneuroim.2016.04.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 12/20/2022]
|
36
|
Staphylococcus aureus Phenol-Soluble Modulins Impair Interleukin Expression in Bovine Mammary Epithelial Cells. Infect Immun 2016; 84:1682-1692. [PMID: 27001539 DOI: 10.1128/iai.01330-15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 03/13/2016] [Indexed: 12/27/2022] Open
Abstract
The role of the recently described interleukin-32 (IL-32) in Staphylococcus aureus-induced mastitis, an inflammation of the mammary gland, is unclear. We determined expression of IL-32, IL-6, and IL-8 in S. aureus- and Escherichia coli-infected bovine mammary gland epithelial cells. Using live bacteria, we found that in S. aureus-infected cells, induction of IL-6 and IL-8 expression was less pronounced than in E. coli-infected cells. Notably, IL-32 expression was decreased in S. aureus-infected cells, while it was increased in E. coli-infected cells. We identified the staphylococcal phenol-soluble modulin (PSM) peptides as key contributors to these effects, as IL-32, IL-6, and IL-8 expression by epithelial cells exposed to psm mutant strains was significantly increased compared to that in cells exposed to the isogenic S. aureus wild-type strain, indicating that PSMs inhibit the production of these interleukins. The use of genetically complemented strains confirmed this observation. Inasmuch as the decreased expression of IL-32, which is involved in dendritic cell maturation, impairs immune responses, our results support a PSM-dependent mechanism that allows for the development of chronic S. aureus-related mastitis.
Collapse
|
37
|
Nicholl MB, Chen X, Qin C, Bai Q, Zhu Z, Davis MR, Fang Y. IL-32α has differential effects on proliferation and apoptosis of human melanoma cell lines. J Surg Oncol 2016; 113:364-9. [DOI: 10.1002/jso.24142] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 12/08/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Michael B. Nicholl
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
- South Texas Veterans Health Care System; San Antonio Texas
| | - Xuhui Chen
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
- Luohu Hospital; Shenzhen China
| | - Chenglu Qin
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
- Luohu Hospital; Shenzhen China
| | - Qian Bai
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
| | - Ziwen Zhu
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
| | - Matthew R. Davis
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
| | - Yujiang Fang
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
- Des Moines University; Des Moines Iowa
| |
Collapse
|
38
|
Shimizu Y, Yamamoto M, Yajima H, Suzuki C, Naishiro Y, Takahashi H, Imai K, Shinomura Y. Role of interleukin-32 in the mechanism of chronic inflammation in IgG4-related disease and as a predictive biomarker for drug-free remission. Mod Rheumatol 2015; 26:391-7. [DOI: 10.3109/14397595.2015.1089973] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Yui Shimizu
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan and
| | - Motohisa Yamamoto
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan and
| | - Hidetaka Yajima
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan and
| | - Chisako Suzuki
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan and
| | - Yasuyoshi Naishiro
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan and
| | - Hiroki Takahashi
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan and
| | - Kohzoh Imai
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yasuhisa Shinomura
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan and
| |
Collapse
|
39
|
Cho KS, Lee EJ, Kim JN, Choi JW, Kim HY, Han SH, Ryu JH, Cheong JH, Shin CY, Kwon KJ. Proteinase 3 Induces Neuronal Cell Death Through Microglial Activation. Neurochem Res 2015; 40:2242-51. [PMID: 26349766 DOI: 10.1007/s11064-015-1714-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/30/2015] [Accepted: 08/28/2015] [Indexed: 01/29/2023]
Abstract
Proteinase 3 (PR3) is released from neutrophil granules and is involved in the inflammatory process. PR3 is implicated in antimicrobial defense and cell death, but the exact role of PR3 in the brain is less defined. Microglia is the major immune effector cells in the CNS and is activated by brain injury. In the present study, the effect of PR3 on glial activation was investigated. Microglial activation was assessed by the intracellular level of reactive oxygen species and expression of inflammatory cytokines. The conditioned media from activated microglia by PR3 was used for measuring the neurotoxic effects of PR3-stimulated microglia. The effects of PR3 in vivo were measured by microinjecting PR3 into the rat brain. Herein we show that PR3 increased the inflammatory responses including the intracellular ROS and pro-inflammatory cytokine production in rat primary microglia. Conditioned media from PR3-treated microglia induced neuronal cell death in a concentration dependent manner. Furthermore, microinjected PR3 into the striatum of the rat brain induced microglial activation and neuronal cell death. Interestingly treatment with anti-PR3 monoclonal antibody and protease inhibitors ameliorated microglial activation induced by PR3 in primary microglia and striatum, which also prevented neuronal cell death in both conditions. The data presented here suggest that PR3 is a direct modulator of microglial activation and causes neuronal death through the augmentation of inflammatory responses. We suggest that PR3 could be a new modulator of neuroinflammation, and blocking PR3 would be a promising novel therapeutic target for neuroinflammatory disease such as stroke and Alzheimer's disease.
Collapse
Affiliation(s)
- Kyu Suk Cho
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, 120 Neungdong-ro, Gwangjin-gu, Seoul, 143-701, Korea
| | - Eun Joo Lee
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, 120 Neungdong-ro, Gwangjin-gu, Seoul, 143-701, Korea
| | - Jung Nam Kim
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, 120 Neungdong-ro, Gwangjin-gu, Seoul, 143-701, Korea
| | - Ji Woong Choi
- Department of Pharmacology, College of Pharmacy, Gachon University, Incheon, Korea
| | - Hahn Young Kim
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, 120 Neungdong-ro, Gwangjin-gu, Seoul, 143-701, Korea.,Department of Neurology, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Seoul, Korea
| | - Seol-Heui Han
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, 120 Neungdong-ro, Gwangjin-gu, Seoul, 143-701, Korea.,Department of Neurology, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Seoul, Korea
| | - Jong Hoon Ryu
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Jae Hoon Cheong
- Department of Pharmacy, College of Pharmacy, Sahmyook University, Seoul, Korea
| | - Chan Young Shin
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, 120 Neungdong-ro, Gwangjin-gu, Seoul, 143-701, Korea
| | - Kyoung Ja Kwon
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, 120 Neungdong-ro, Gwangjin-gu, Seoul, 143-701, Korea.
| |
Collapse
|
40
|
Bai X, Dinarello CA, Chan ED. The role of interleukin-32 against tuberculosis. Cytokine 2015; 76:585-587. [PMID: 26144292 DOI: 10.1016/j.cyto.2015.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 12/12/2022]
Abstract
IL-32 is increasingly recognized to be an important host-protective molecule against tuberculosis. In this commentary, we highlight some of the potential mechanisms by which the immunomodulatory effect of IL-32 occurs against mycobacterial infections but also areas where mechanistic clarifications are needed.
Collapse
Affiliation(s)
- Xiyuan Bai
- Denver Veterans Affairs Medical Center, 1055 Clermont St, Denver, CO 80206, United States; Departments of Medicine and Academic Affairs, D509, Neustadt Building, National Jewish Health, 1400 Jackson St, Denver, CO 80206, United States; Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, 12700 East 19th Avenue, Aurora, CO 80045-2539, United States
| | - Charles A Dinarello
- Division of Infectious Diseases, University of Colorado Denver Anschutz Medical Campus, 12700 East 19th Avenue, Aurora, CO 80045-2539, United States
| | - Edward D Chan
- Denver Veterans Affairs Medical Center, 1055 Clermont St, Denver, CO 80206, United States; Departments of Medicine and Academic Affairs, D509, Neustadt Building, National Jewish Health, 1400 Jackson St, Denver, CO 80206, United States; Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, 12700 East 19th Avenue, Aurora, CO 80045-2539, United States.
| |
Collapse
|
41
|
Li D, Chen D, Zhang X, Wang H, Song Z, Xu W, He Y, Yin Y, Cao J. c-Jun N-terminal kinase and Akt signalling pathways regulating tumour necrosis factor-α-induced interleukin-32 expression in human lung fibroblasts: implications in airway inflammation. Immunology 2015; 144:282-90. [PMID: 25157456 DOI: 10.1111/imm.12374] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 12/22/2022] Open
Abstract
Airway inflammatory diseases such as chronic obstructive pulmonary disease (COPD) and asthma are associated with elevated expression of interleukin-32 (IL-32), a recently described cytokine that appears to play a critical role in inflammation. However, so far, the regulation of pulmonary IL-32 production has not been fully established. We examined the expression of IL-32 by tumour necrosis factor-α (TNF-α) in primary human lung fibroblasts. Human lung fibroblasts were cultured in the presence or absence of TNF-α and/or other cytokines/Toll-like receptor (TLR) ligands or various signalling molecule inhibitors to analyse the expression of IL-32 by quantitative RT-PCR and ELISA. Next, activation of Akt and c-Jun N-terminal kinase (JNK) signalling pathways was investigated by Western blot. Interleukin-32 mRNA of four spliced isoforms (α, β, γ and δ) was up-regulated upon TNF-α stimulation, which was associated with a significant IL-32 protein release from TNF-α-activated human lung fibroblasts. The combination of interferon-γ and TNF-α induced enhanced IL-32 release in human lung fibroblasts, whereas IL-4, IL-17A, IL-27 and TLR ligands did not alter IL-32 release in human lung fibroblasts either alone, or in combination with TNF-α. Furthermore, the activation of Akt and JNK pathways regulated TNF-α-induced IL-32 expression in human lung fibroblasts, and inhibition of the Akt and JNK pathways was able to suppress the increased release of IL-32 to nearly the basal level. These data suggest that TNF-α may be involved in airway inflammation via the induction of IL-32 by activating Akt and JNK signalling pathways. Therefore, the TNF-α/IL-32 axis may be a potential therapeutic target for airway inflammatory diseases.
Collapse
Affiliation(s)
- Dagen Li
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Leonurine ameliorates kidney fibrosis via suppressing TGF-β and NF-κB signaling pathway in UUO mice. Int Immunopharmacol 2015; 25:406-15. [DOI: 10.1016/j.intimp.2015.02.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 02/09/2015] [Accepted: 02/12/2015] [Indexed: 12/29/2022]
|
43
|
Monteleone K, Selvaggi C, Cacciotti G, Falasca F, Mezzaroma I, D'Ettorre G, Turriziani O, Vullo V, Antonelli G, Scagnolari C. MicroRNA-29 family expression and its relation to antiviral immune response and viro-immunological markers in HIV-1-infected patients. BMC Infect Dis 2015; 15:51. [PMID: 25808800 PMCID: PMC4336718 DOI: 10.1186/s12879-015-0768-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 01/16/2015] [Indexed: 12/22/2022] Open
Abstract
Background Several in vitro studies suggested the microRNA-29 (miRNA-29) family is involved in regulating HIV-1 and modulating the expression of interleukin (IL)-32, an anti-HIV-1 cytokine. Methods To investigate the contribution of the miRNA-29 family to HIV-1 infection in vivo, we compared miRNA-29 expression in PBMC collected from 58 HIV-1-infected patients, naïve for antiretroviral therapy, and 21 gender- and age-matched HIV-1 seronegative healthy donors, using RT-Taqman assays. The relation between miRNA-29 levels and HIV-1 viro-immunological markers and the activation rate of antiviral immune response were also evaluated. In addition, we profiled miRNA-29 expression in CD4+ T lymphocytes and CD14+ monocytes collected from 5 antiretroviral treated HIV-1 infected patients. Results miRNA-29b levels were higher in HIV-1-infected patients than in the control group (p < 0.001). There were no correlations with either HIV-1 RNA levels or CD4+ T count, whereas a significant correlation was found between miRNA-29-a/c levels and integrated HIV-1 DNA (miRNA-29a: p = 0.009, r = −0.448; miRNA-29c: p = 0.029; r = −0.381). When the HIV-1-infected patients were grouped on the basis of their plasma HIV-1 RNA and CD4+ T cell count, we also found that patients expressing the lowest levels of miRNA-29c showed high viraemia, low CD4+ T cell count and high levels of integrated HIV-1 DNA. Moreover, miRNA-29b levels were correlated with those of IL-32nonα (p = 0.028; r = −0.298). Patients expressing higher levels of miRNA-29b showed lower levels of MxA, an interferon-stimulated gene, also induced by IL-32 (p = 0.006 r = −0.397). Lastly, we found that CD4+ T lymphocytes and CD14+ monocytes shared similar miRNA-29a/b/c expression patterns but the amount of miRNA-29a/b/c, IL-32 isoforms and MxA were highly variable in these two cellular subsets. Conclusions The miRNA-29 family could influence the clinical progression of HIV-1 infection, the HIV-1 proviral load and the innate immune response against HIV-1.
Collapse
|
44
|
Allaeys I, Gymninova I, Canet-Jourdan C, Poubelle PE. IL-32γ delays spontaneous apoptosis of human neutrophils through MCL-1, regulated primarily by the p38 MAPK pathway. PLoS One 2014; 9:e109256. [PMID: 25275312 PMCID: PMC4183585 DOI: 10.1371/journal.pone.0109256] [Citation(s) in RCA: 6] [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: 05/25/2014] [Accepted: 09/09/2014] [Indexed: 12/21/2022] Open
Abstract
IL-32γ is a multifunctional cytokine involved in various inflammatory and auto-immune diseases in which neutrophils can affect the evolution of these diseases. To persist at inflammatory sites, neutrophils require inhibition of their rapid and constitutive apoptosis, an inhibitory effect that phlogogenic cytokines support. To date, the effects of IL-32γ on neutrophils remain unknown. We demonstrate that IL-32γ delays, in a dose-dependent manner, the spontaneous apoptosis of human blood neutrophils by activating mainly p38 MAPK through rapid p38 phosphorylation. PI3-K and ERK1/2 MAPK are also involved, but to a lesser extent. Most of cytokines that induce retardation of neutrophil apoptosis activate the expression of MCL-1 at both mRNA and protein levels. IL-32γ added to human blood neutrophils in vitro is associated with sustained levels of MCL-1 protein. This effect in neutrophils corresponds to a decrease of MCL-1 protein degradation without any effect on MCL-1 mRNA levels. The sustained levels of MCL-1 induced by IL-32γ are only abrogated by the p38β MAPK inhibitor SB202190. Additionally, IL-32γ induces a reduction in caspase 3 activity in neutrophils. In conclusion, IL-32γ affects human blood neutrophils in vitro by increasing their survival, suggesting that this cytokine could have profound effects on the deleterious functions of neutrophils in several diseases.
Collapse
Affiliation(s)
- Isabelle Allaeys
- Centre de Recherche en Rhumatologie et Immunologie (CRRI), Centre de Recherche du CHU de Québec, Département de Médecine, Université Laval, Québec, Canada
| | - Irina Gymninova
- Centre de Recherche en Rhumatologie et Immunologie (CRRI), Centre de Recherche du CHU de Québec, Département de Médecine, Université Laval, Québec, Canada
| | - Charlotte Canet-Jourdan
- Centre de Recherche en Rhumatologie et Immunologie (CRRI), Centre de Recherche du CHU de Québec, Département de Médecine, Université Laval, Québec, Canada
| | - Patrice E. Poubelle
- Centre de Recherche en Rhumatologie et Immunologie (CRRI), Centre de Recherche du CHU de Québec, Département de Médecine, Université Laval, Québec, Canada
- * E-mail:
| |
Collapse
|
45
|
Park YS, Kang JW, Lee DH, Kim MS, Bak Y, Yang Y, Lee HG, Hong J, Yoon DY. Interleukin-32α modulates promyelocytic leukemia zinc finger gene activity by inhibiting protein kinase Cɛ-dependent sumoylation. Int J Biochem Cell Biol 2014; 55:136-43. [DOI: 10.1016/j.biocel.2014.08.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 07/17/2014] [Accepted: 08/21/2014] [Indexed: 01/20/2023]
|
46
|
Xu D, Chen M, Ren X, Ren X, Wu Y. Leonurine ameliorates LPS-induced acute kidney injury via suppressing ROS-mediated NF-κB signaling pathway. Fitoterapia 2014; 97:148-55. [DOI: 10.1016/j.fitote.2014.06.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/01/2014] [Accepted: 06/02/2014] [Indexed: 01/22/2023]
|
47
|
Snelgrove RJ, Gregory LG, Peiró T, Akthar S, Campbell GA, Walker SA, Lloyd CM. Alternaria-derived serine protease activity drives IL-33-mediated asthma exacerbations. J Allergy Clin Immunol 2014; 134:583-592.e6. [PMID: 24636086 PMCID: PMC4152000 DOI: 10.1016/j.jaci.2014.02.002] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 01/29/2014] [Accepted: 02/03/2014] [Indexed: 12/24/2022]
Abstract
Background The fungal allergen Alternaria alternata is implicated in severe asthma and rapid onset life-threatening exacerbations of disease. However, the mechanisms that underlie this severe pathogenicity remain unclear. Objective We sought to investigate the mechanism whereby Alternaria was capable of initiating severe, rapid onset allergic inflammation. Methods IL-33 levels were quantified in wild-type and ST2−/− mice that lacked the IL-33 receptor given inhaled house dust mite, cat dander, or Alternaria, and the effect of inhibiting allergen-specific protease activities on IL-33 levels was assessed. An exacerbation model of allergic airway disease was established whereby mice were sensitized with house dust mite before subsequently being challenged with Alternaria (with or without serine protease activity), and inflammation, remodeling, and lung function assessed 24 hours later. Results Alternaria, but not other common aeroallergens, possessed intrinsic serine protease activity that elicited the rapid release of IL-33 into the airways of mice through a mechanism that was dependent upon the activation of protease activated receptor-2 and adenosine triphosphate signaling. The unique capacity of Alternaria to drive this early IL-33 release resulted in a greater pulmonary inflammation by 24 hours after challenge relative to the common aeroallergen house dust mite. Furthermore, this Alternaria serine protease–IL-33 axis triggered a rapid, augmented inflammation, mucus release, and loss of lung function in our exacerbation model. Conclusion Alternaria-specific serine protease activity causes rapid IL-33 release, which underlies the development of a robust TH2 inflammation and exacerbation of allergic airway disease.
Collapse
Affiliation(s)
- Robert J Snelgrove
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Lisa G Gregory
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Teresa Peiró
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Samia Akthar
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Gaynor A Campbell
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Simone A Walker
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Clare M Lloyd
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
48
|
Monteleone K, Di Maio P, Cacciotti G, Falasca F, Fraulo M, Falciano M, Mezzaroma I, D'Ettorre G, Turriziani O, Scagnolari C. Interleukin-32 isoforms: expression, interaction with interferon-regulated genes and clinical significance in chronically HIV-1-infected patients. Med Microbiol Immunol 2014; 203:207-16. [PMID: 24553842 DOI: 10.1007/s00430-014-0329-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 01/28/2014] [Indexed: 11/25/2022]
Abstract
Given the growing evidence for a role of interleukin-32 (IL-32) in the immune response to HIV-1 infection and its interplay with type I and III interferons (IFNs), we studied the gene expression of IL-32 isoforms (α and nonα) in untreated chronically HIV-1-infected patients and in gender- and age-matched healthy individuals. To further characterize both the anti-HIV properties of IL-32 and the cytokine's relationship with host antiviral innate immune responses, we evaluated whether IL-32 can induce ex vivo the expression of antiviral IFN-induced genes (ISGs), namely myxovirus resistance A (MxA), and apolipoprotein B mRNA-editing enzyme catalytic (APOBEC)3G and APOBEC3F. We also investigated whether in vivo IL-32 (α and nonα) mRNA levels were correlated with those of MxA and APOBEC3G/3F. Results indicated that IL-32 (α and nonα) mRNA levels were significantly higher in HIV-1-infected patients than in healthy individuals. Furthermore, IL-32 (α and nonα) mRNA levels correlated negatively with HIV RNA levels, but not with the CD4(+) T-cell count. Our ex vivo studies disclosed that ISGs mRNA levels were increased after IL-32γ treatment of peripheral blood mononuclear cells. Interestingly, significant positive correlations were found between transcript levels of both IL-32α and IL-32nonα and those of MxA and APOBEC3G/3F in untreated chronically HIV-1-infected patients. Overall, our results demonstrated that IL-32 isoforms are highly expressed during chronic HIV-1 infection and that IL-32 could have a central role in the antiviral immune response against HIV-1.
Collapse
Affiliation(s)
- Katia Monteleone
- Pasteur Institute-Cenci Bolognetti Foundation, Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Viale di Porta Tiburtina n 28, 00185, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|