1
|
Mester P, Utrata A, Schmidtner N, Birner C, Schmid S, Müller M, Pavel V, Buechler C. Lower Plasma IL-32 Levels Linked to Better Survival in Sepsis. Biomedicines 2025; 13:750. [PMID: 40149726 PMCID: PMC11940173 DOI: 10.3390/biomedicines13030750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Interleukin-32 (IL-32) is a pro-inflammatory cytokine primarily produced by immune cells and involved in bacterial and viral infections. This study investigates whether plasma IL-32 is associated with sepsis severity and clinical outcomes. Methods: Plasma IL-32 levels were measured in 186 patients with systemic inflammatory response syndrome (SIRS), sepsis, or septic shock, as well as in 40 controls. The relationship between IL-32 levels and SARS-CoV-2 or bacterial infections, alongside underlying etiological conditions, was assessed. Results: Patients with liver cirrhosis exhibited elevated plasma IL-32 levels. After excluding these patients, IL-32 levels were lower in SIRS/sepsis patients compared to the controls. No significant differences in IL-32 levels were observed among SIRS, sepsis, and septic shock patients. Additionally, underlying conditions such as pancreatitis and cholangitis did not influence IL-32 levels. Patients with bloodstream bacterial infections, SARS-CoV-2 infections, or no documented infection had comparable IL-32 levels. Notably, higher IL-32 levels were associated with increased mortality. Conclusions: These findings suggest that a reduction in plasma IL-32 levels may be protective in SIRS/sepsis patients, as elevated levels are linked to poor survival outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christa Buechler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (P.M.); (A.U.); (N.S.); (C.B.); (S.S.); (M.M.); (V.P.)
| |
Collapse
|
2
|
Gniadecki R, Osman M, Hennesey D, O'Keefe S, Thomsen SF, Iyer A. Architecture of skin inflammation in psoriasis revealed by spatial transcriptomics. Clin Immunol 2023; 256:109771. [PMID: 37708923 DOI: 10.1016/j.clim.2023.109771] [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: 06/28/2023] [Revised: 07/24/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023]
Abstract
Psoriasis is a chronic inflammatory skin disease, thought to be predominantly mediated by TH17 cells. Significance of other inflammatory pathways and the innate immune system is not well understood and the spatial heterogeneity of inflammation in the skin has largely been overlooked. Our aim was to create a comprehensive map of skin inflammation in psoriasis, exploring the tissue patterning of inflammation. In situ whole transcriptome sequencing (spatial sequencing) was performed on lesional psoriatic skin in four patients with moderate-to-severe disease to quantify all expressed genes within a tissue section. Transcriptional analysis revealed three major inflammatory niches in psoriasis skin, each with distinct cytokine circuits and chemokines: the hyperplastic epidermis, upper (papillary) dermis, and reticular dermis. Interestingly, key cytokines such as IL-23, IL-17 s, and TNFα were not notably present in the skin's transcriptomic signature. Unexpectedly, IL-32 showed strong expression in the dermis. Our findings underscore the complexity of psoriatic inflammation, highlighting its architectural heterogeneity and the roles of innate cytokines. Both IL-32 and IL-1 family cytokines appear to play critical roles in the dermal and epidermal inflammation, respectively, and may provide pharmacological targets to improve the control of the inflammatory process.
Collapse
Affiliation(s)
- Robert Gniadecki
- Division of Dermatology, Department of Medicine, University of Alberta, Canada.
| | - Mohammed Osman
- Division of Rheumatology, Department of Medicine, University of Alberta, Canada
| | - Dylan Hennesey
- Division of Dermatology, Department of Medicine, University of Alberta, Canada
| | - Sandra O'Keefe
- Division of Dermatology, Department of Medicine, University of Alberta, Canada
| | | | - Aishwarya Iyer
- Division of Dermatology, Department of Medicine, University of Alberta, Canada
| |
Collapse
|
3
|
Nappi F, Alzamil A, Avtaar Singh SS, Spadaccio C, Bonnet N. Current Knowledge on the Interaction of Human Cytomegalovirus Infection, Encoded miRNAs, and Acute Aortic Syndrome. Viruses 2023; 15:2027. [PMID: 37896804 PMCID: PMC10611417 DOI: 10.3390/v15102027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Aortic dissection is a clinicopathological entity caused by rupture of the intima, leading to a high mortality if not treated. Over time, diagnostic and investigative methods, antihypertensive therapy, and early referrals have resulted in improved outcomes according to registry data. Some data have also emerged from recent studies suggesting a link between Human Cytomegalovirus (HCMV) infection and aortic dissection. Furthermore, the use of microRNAs has also become increasingly widespread in the literature. These have been noted to play a role in aortic dissections with elevated levels noted in studies as early as 2017. This review aims to provide a broad and holistic overview of the role of miRNAs, while studying the role of HCMV infection in the context of aortic dissections. The roles of long non-coding RNAs, circular RNAs, and microRNAs are explored to identify changes in expression during aortic dissections. The use of such biomarkers may one day be translated into clinical practice to allow early detection and prognostication of outcomes and drive preventative and therapeutic options in the future.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (A.A.); (N.B.)
| | - Almothana Alzamil
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (A.A.); (N.B.)
| | | | - Cristiano Spadaccio
- Department of Cardiothoracic Surgery, Mayo Clinic, Rochester, Rochester, MN 55905, USA;
| | - Nicolas Bonnet
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (A.A.); (N.B.)
| |
Collapse
|
4
|
Cross AR, de Andrea CE, Villalba-Esparza M, Landecho MF, Cerundolo L, Weeratunga P, Etherington RE, Denney L, Ogg G, Ho LP, Roberts IS, Hester J, Klenerman P, Melero I, Sansom SN, Issa F. Spatial transcriptomic characterization of COVID-19 pneumonitis identifies immune circuits related to tissue injury. JCI Insight 2023; 8:e157837. [PMID: 36472908 PMCID: PMC9977306 DOI: 10.1172/jci.insight.157837] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Severe lung damage resulting from COVID-19 involves complex interactions between diverse populations of immune and stromal cells. In this study, we used a spatial transcriptomics approach to delineate the cells, pathways, and genes present across the spectrum of histopathological damage in COVID-19-affected lung tissue. We applied correlation network-based approaches to deconvolve gene expression data from 46 areas of interest covering more than 62,000 cells within well-preserved lung samples from 3 patients. Despite substantial interpatient heterogeneity, we discovered evidence for a common immune-cell signaling circuit in areas of severe tissue that involves crosstalk between cytotoxic lymphocytes and pro-inflammatory macrophages. Expression of IFNG by cytotoxic lymphocytes was associated with induction of chemokines, including CXCL9, CXCL10, and CXCL11, which are known to promote the recruitment of CXCR3+ immune cells. The TNF superfamily members BAFF (TNFSF13B) and TRAIL (TNFSF10) were consistently upregulated in the areas with severe tissue damage. We used published spatial and single-cell SARS-CoV-2 data sets to validate our findings in the lung tissue from additional cohorts of patients with COVID-19. The resulting model of severe COVID-19 immune-mediated tissue pathology may inform future therapeutic strategies.
Collapse
Affiliation(s)
- Amy R. Cross
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | | | | | - Manuel F. Landecho
- Department of Internal Medicine, and
- Department of Immunology and Immunotherapy, Clínica de la Universidad de Navarra, Pamplona, Spain
| | - Lucia Cerundolo
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Praveen Weeratunga
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Rachel E. Etherington
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Laura Denney
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Graham Ogg
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Ling-Pei Ho
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Ian S.D. Roberts
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Joanna Hester
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ignacio Melero
- Department of Immunology and Immunotherapy, Clínica de la Universidad de Navarra, Pamplona, Spain
- CIBERONC, Madrid, Spain
- Center for Applied Medical Research, Pamplona, Spain
| | - Stephen N. Sansom
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
5
|
Bowles KR, Pugh DA, Liu Y, Patel T, Renton AE, Bandres-Ciga S, Gan-Or Z, Heutink P, Siitonen A, Bertelsen S, Cherry JD, Karch CM, Frucht SJ, Kopell BH, Peter I, Park YJ, Charney A, Raj T, Crary JF, Goate AM. 17q21.31 sub-haplotypes underlying H1-associated risk for Parkinson's disease are associated with LRRC37A/2 expression in astrocytes. Mol Neurodegener 2022; 17:48. [PMID: 35841044 PMCID: PMC9284779 DOI: 10.1186/s13024-022-00551-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 06/21/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is genetically associated with the H1 haplotype of the MAPT 17q.21.31 locus, although the causal gene and variants underlying this association have not been identified. METHODS To better understand the genetic contribution of this region to PD and to identify novel mechanisms conferring risk for the disease, we fine-mapped the 17q21.31 locus by constructing discrete haplotype blocks from genetic data. We used digital PCR to assess copy number variation associated with PD-associated blocks, and used human brain postmortem RNA-seq data to identify candidate genes that were then further investigated using in vitro models and human brain tissue. RESULTS We identified three novel H1 sub-haplotype blocks across the 17q21.31 locus associated with PD risk. Protective sub-haplotypes were associated with increased LRRC37A/2 copy number and expression in human brain tissue. We found that LRRC37A/2 is a membrane-associated protein that plays a role in cellular migration, chemotaxis and astroglial inflammation. In human substantia nigra, LRRC37A/2 was primarily expressed in astrocytes, interacted directly with soluble α-synuclein, and co-localized with Lewy bodies in PD brain tissue. CONCLUSION These data indicate that a novel candidate gene, LRRC37A/2, contributes to the association between the 17q21.31 locus and PD via its interaction with α-synuclein and its effects on astrocytic function and inflammatory response. These data are the first to associate the genetic association at the 17q21.31 locus with PD pathology, and highlight the importance of variation at the 17q21.31 locus in the regulation of multiple genes other than MAPT and KANSL1, as well as its relevance to non-neuronal cell types.
Collapse
Affiliation(s)
- Kathryn R. Bowles
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Derian A. Pugh
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Yiyuan Liu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Tulsi Patel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Alan E. Renton
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Sara Bandres-Ciga
- Laboratory of Neurogenetics, National Institute On Aging, National Institutes of Health, Bethesda, MD USA
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, Québec Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, Québec Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec Canada
| | - Peter Heutink
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Ari Siitonen
- Institute of Clinical Medicine, Department of Neurology, University of Oulu, Oulu, Finland
- Department of Neurology and Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Sarah Bertelsen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Jonathan D. Cherry
- Alzheimer’s Disease and CTE Center, Boston University, Boston University School of Medicine, Boston, MA USA
- Department of Neurology, Boston University School of Medicine, Boston, MA USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA USA
| | - Celeste M. Karch
- Department of Psychiatry, Washington University in St Louis, St. Louis, MO USA
| | - Steven J. Frucht
- Department of Neurology, Fresco Institute for Parkinson’s and Movement Disorders, New York University Langone, New York, NY USA
| | - Brian H. Kopell
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Center for Neuromodulation, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Inga Peter
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Y. J. Park
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | | | - Alexander Charney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Towfique Raj
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - John F. Crary
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - A. M. Goate
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| |
Collapse
|
6
|
Shim S, Lee S, Hisham Y, Kim S, Nguyen TT, Taitt AS, Hwang J, Jhun H, Park HY, Lee Y, Yeom SC, Kim SY, Kim YG, Kim S. Comparison of the Seven Interleukin-32 Isoforms’ Biological Activities: IL-32θ Possesses the Most Dominant Biological Activity. Front Immunol 2022; 13:837588. [PMID: 35281066 PMCID: PMC8914309 DOI: 10.3389/fimmu.2022.837588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Cytokines are significantly associated with the homeostasis of immune responses in health and disease. Interleukin-32 (IL-32) is a cytokine originally discovered in natural killer cell transcript 4. IL-32 with different disorders has been described in terms of pathogenesis and the progression of diseases. Clinical studies have investigated IL-32 under various conditions, such as viral infection, autoimmune diseases, inflammatory diseases, certain types of cancer, vascular disease, and pulmonary diseases. The high expression of IL-32 was identified in different tissues with various diseases and found to have multiple transcripts of up to seven isoforms. However, the purification and biological activities of these isoforms have not been investigated yet. Therefore, in this study, we purified and compared the biological activity of recombinant IL-32 (rIL-32) isoforms. This is the first time for seven rIL-32 isoforms (α, β, δ, γ, ϵ, ζ, and θ) to be cloned and purified using an Escherichia coli expression system. Next, we evaluate the biological activities of these seven rIL-32 isoforms, which were used to treat different types of cells by assessing the levels of inflammatory cytokine production. The results revealed that rIL-32θ possessed the most dominant biological activity in both immune and non-immune cells.
Collapse
Affiliation(s)
- Saerok Shim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Siyoung Lee
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Yasmin Hisham
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Sinae Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
- College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Tam T. Nguyen
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
- College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Afeisha S. Taitt
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Jihyeong Hwang
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Hyunjhung Jhun
- Technical Assistance Center, Korea Food Research Institute, Wanju, South Korea
| | - Ho-Young Park
- Research Group of Functional Food Materials, Korea Food Research Institute, Wanju, South Korea
| | - Youngmin Lee
- Department of Medicine, Pusan Paik Hospital, Collage of Medicine, Inje University, Busan, South Korea
| | - Su Cheong Yeom
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, South Korea
| | - Sang-Yeob Kim
- Convergence Medicine Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul, South Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- *Correspondence: Yong-Gil Kim, ; Soohyun Kim,
| | - Soohyun Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
- College of Veterinary Medicine, Konkuk University, Seoul, South Korea
- *Correspondence: Yong-Gil Kim, ; Soohyun Kim,
| |
Collapse
|
7
|
Hatatian N, Bosstani R, Mohammadi A, Mehraban S, Mahdifar M, Zemorshidi F, Mozhgani SH, Haji Ghadimi A, Foroughipour M, Rafatpanah H. Evaluation of interleukin-32 and cyclooxygenase-2 expression in HAM/TSP patients and HTLV-1 asymptomatic carriers. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:992-996. [PMID: 34712431 PMCID: PMC8528256 DOI: 10.22038/ijbms.2021.50821.11569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 06/07/2021] [Indexed: 11/13/2022]
Abstract
Objective(s): HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a neuroinflammatory disorder associated with HTLV-1. Cytokines and inflammatory mediators have a major role in forming inflammation in HAM/TSP patients. This study aimed to measure the levels of IL-32, a proinflammatory cytokine associated with autoinflammatory disorders, and also cyclooxygenase -2 (COX-2) as a key mediator of inflammatory pathways in HAM/TSP patients and HTLV-1 asymptomatic carriers (ACs). Materials and Methods: Peripheral blood monocyte cells (PBMCs) were isolated from HAM/TSP patients, ACs, and healthy controls (HCs), and DNA and RNA were extracted to evaluate HTLV-1 proviral load (PVL) and expression of IL-32 and COX-2, using real-time PCR. Serum levels of IL-32 were determined by using an ELISA assay. Results: The expression level of IL-32 was significantly higher in ACs compared with HAM/TSP patients and HCs (P<0.0001 and P>0.05, respectively). There were no statistically significant differences in the expression levels of Cox-2 and protein levels of IL-32 between the study groups. HTLV-1 PVL was higher in HAM/TSP patients compared with ACs. Conclusion: Results showed increased mRNA levels of IL-32 in ACs. Since HTLV-1 PVL in ACs is lower than in HAM/TSP patients, it could be concluded that IL-32 might be an HTLV-1 inhibitor that seems to control virus replication. Despite the difference in IL-32 mRNA levels between study groups, no statistically significant differences were observed in IL-32 serum levels. Also, there were no significant differences in COX-2 expression.
Collapse
Affiliation(s)
- Niayesh Hatatian
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Bosstani
- Department of Neurology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asadollah Mohammadi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Saeedeh Mehraban
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Mahdifar
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fariba Zemorshidi
- Department of Neurology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sayed-Hamidreza Mozhgani
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Mohsen Foroughipour
- Department of Neurology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Gabriel EM, Wiche Salinas TR, Gosselin A, Larouche-Anctil E, Durand M, Landay AL, El-Far M, Tremblay CL, Routy JP, Ancuta P. Overt IL-32 isoform expression at intestinal level during HIV-1 infection is negatively regulated by IL-17A. AIDS 2021; 35:1881-1894. [PMID: 34101628 PMCID: PMC8416712 DOI: 10.1097/qad.0000000000002972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Untreated HIV infection was previously associated with IL-32 overexpression in gut/intestinal epithelial cells (IEC). Here, we explored IL-32 isoform expression in the colon of people with HIV (PWH) receiving antiretroviral therapy (ART) and IL-32 triggers/modulators in IEC. DESIGN Sigmoid colon biopsies (SCB) and blood were collected from ART-treated PWH (HIV + ART; n = 17; mean age: 56 years; CD4+ cell counts: 679 cells/μl; time on ART: 72 months) and age-matched HIV-uninfected controls (HIVneg; n = 5). The IEC line HT-29 was used for mechanistic studies. METHODS Cells from SCB and blood were isolated by enzymatic digestion and/or gradient centrifugation. HT-29 cells were exposed to TLR1-9 agonists, TNF-α, IL-17A and HIV. IL-32α/β/γ/D/ε/θ and IL-17A mRNA levels were quantified by real-time RT-PCR. IL-32 protein levels were quantified by ELISA. RESULTS IL-32β/γ/ε isoform transcripts were detectable in the blood and SCB, with IL-32β mRNA levels being predominantly expressed in both compartments and at significantly higher levels in HIV + ART compared to HIVneg. IL-17A transcripts were only detectable in SCB, with increased IL-17A levels in HIVneg compared with HIV + ART and negatively correlated with IL-32β mRNA levels. IL-32β/γ/ε isoform mRNA were detected in HT-29 cells upon exposure to TNF-α, Poly I:C (TLR3 agonist), Flagellin (TLR-5 agonist) and HIV. IL-17A significantly decreased both IL-32 β/γ/ε mRNA and cell-associated IL-32 protein levels induced upon TNF-α and Poly I:C triggering. CONCLUSION We document IL-32 isoforms abundant in the colon of ART-treated PWH and reveal the capacity of the Th17 hallmark cytokine IL-17A to attenuate IL-32 overexpression in a model of inflamed IEC.
Collapse
Affiliation(s)
- Etiene Moreira Gabriel
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Tomas Raul Wiche Salinas
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | | | | | - Madeleine Durand
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | | | | | - Cécile L. Tremblay
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Pierre Routy
- McGill University Health Centre, Montreal, Québec, Canada
- Chronic Viral Illness Service and Hematology Department, McGill University Health Centre, Montréal, Québec, Canada
| | - Petronela Ancuta
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
9
|
Abstract
Regulatory T cells (Tregs) are responsible for restraining excessive inflammation, a hallmark of COVID-19. We identified a striking phenotype in Tregs from patients with severe disease, as well as an interesting role for interleukin (IL)-6 and IL-18. An increased suppressive profile, including increased Treg proportions, combined with the expression of proinflammatory mediators, distinguished severe patients and persisted in some of those recovered. This phenotype is in notable similarity to that found in tumor-infiltrating Tregs, which are generally associated with poor prognosis, and suggests both a detrimental role for these cells in COVID-19 as well as a potential explanation for some of the still largely unexplored complications associated with recovery. The hallmark of severe COVID-19 is an uncontrolled inflammatory response, resulting from poorly understood immunological dysfunction. We hypothesized that perturbations in FoxP3+ T regulatory cells (Treg), key enforcers of immune homeostasis, contribute to COVID-19 pathology. Cytometric and transcriptomic profiling revealed a distinct Treg phenotype in severe COVID-19 patients, with an increase in Treg proportions and intracellular levels of the lineage-defining transcription factor FoxP3, correlating with poor outcomes. These Tregs showed a distinct transcriptional signature, with overexpression of several suppressive effectors, but also proinflammatory molecules like interleukin (IL)-32, and a striking similarity to tumor-infiltrating Tregs that suppress antitumor responses. Most marked during acute severe disease, these traits persisted somewhat in convalescent patients. A screen for candidate agents revealed that IL-6 and IL-18 may individually contribute different facets of these COVID-19–linked perturbations. These results suggest that Tregs may play nefarious roles in COVID-19, by suppressing antiviral T cell responses during the severe phase of the disease, and by a direct proinflammatory role.
Collapse
|
10
|
Wang F, Huang G, Shen L, Peng Y, Sha W, Chen ZW, Shen H. Genetics and Functional Mechanisms of STAT3 Polymorphisms in Human Tuberculosis. Front Cell Infect Microbiol 2021; 11:669394. [PMID: 34307193 PMCID: PMC8294188 DOI: 10.3389/fcimb.2021.669394] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/14/2021] [Indexed: 01/17/2023] Open
Abstract
Signal transducer and activator of transcription-3 (STAT3) plays an important role in biological balance. Our and others previous studies implied that STAT3 had a great effect on fast-acting innate immunity against tuberculosis (TB). We hypothesized that stat3 SNP down-regulation of STAT3 leads to a change in susceptibility to TB in humans. To test this hypothesis, we investigated STAT3 SNPs using SNP scan™ technique in a case-control study of TB patients (n = 470) and HC subjects (n = 356), and then conducted functional studies of them using cellular models. We found that SNPs in STAT3 3`-UTR of rs1053004 TT and rs1053005 AA genotypes or T-A haplotype were associated with susceptibility to TB or TB severity. While the TT/AA genotype correlated with the low constitutive expression of stat3 and IL-17A in PBMC, the variant stat3 of rs1053004-rs1053005 T-A haplotype indeed reduced stat3 expression in reporter assays. Interestingly, host PBMC expressing the rs1053005 AA genotype and low constitutive stat3 exhibited the reduced ability to mount fast-acting innate immunity against mycobacterial infection in cellular models. Finally, mechanistic experiments showed that the STAT3 down-regulation broadly depressed STAT3 downstream anti-mycobacterial activities involving VDR-related CAMP pathway as well as IL-32, iNOS and autophagy mechanisms, leading to an enhanced mycobacterial infection. The findings of this study suggest that low constitutive stat3 derived from the TT/AA genotype/T-A haplotype acts to down-regulate STAT3, depressing multiple anti-mycobacterial pathways/mechanisms downstream, which leads to an enhanced mycobacterial infection or TB in high-risk individuals.
Collapse
Affiliation(s)
- Feifei Wang
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guixian Huang
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Ling Shen
- Department of Microbiology & Immunology and Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, United States
| | - Ying Peng
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Wei Sha
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Zheng W Chen
- Department of Microbiology & Immunology and Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, United States
| | - Hongbo Shen
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Genome-wide bioinformatic analyses predict key host and viral factors in SARS-CoV-2 pathogenesis. Commun Biol 2021; 4:590. [PMID: 34002013 PMCID: PMC8128904 DOI: 10.1038/s42003-021-02095-0] [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] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 04/05/2021] [Indexed: 02/03/2023] Open
Abstract
The novel betacoronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused a worldwide pandemic (COVID-19) after emerging in Wuhan, China. Here we analyzed public host and viral RNA sequencing data to better understand how SARS-CoV-2 interacts with human respiratory cells. We identified genes, isoforms and transposable element families that are specifically altered in SARS-CoV-2-infected respiratory cells. Well-known immunoregulatory genes including CSF2, IL32, IL-6 and SERPINA3 were differentially expressed, while immunoregulatory transposable element families were upregulated. We predicted conserved interactions between the SARS-CoV-2 genome and human RNA-binding proteins such as the heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) and eukaryotic initiation factor 4 (eIF4b). We also identified a viral sequence variant with a statistically significant skew associated with age of infection, that may contribute to intracellular host-pathogen interactions. These findings can help identify host mechanisms that can be targeted by prophylactics and/or therapeutics to reduce the severity of COVID-19.
Collapse
|
12
|
Galvan-Pena S, Leon J, Chowdhary K, Michelson DA, Vijaykumar B, Yang L, Magnuson A, Manickas-Hill Z, Piechocka-Trocha A, Worrall DP, Hall KE, Ghebremichael M, Walker BD, Li JZ, Yu XG, Mathis D, Benoist C. Profound Treg perturbations correlate with COVID-19 severity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 33330871 DOI: 10.1101/2020.12.11.416180] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The hallmark of severe COVID-19 disease has been an uncontrolled inflammatory response, resulting from poorly understood immunological dysfunction. We explored the hypothesis that perturbations in FoxP3+ T regulatory cells (Treg), key enforcers of immune homeostasis, contribute to COVID-19 pathology. Cytometric and transcriptomic profiling revealed a distinct Treg phenotype in severe COVID-19 patients, with an increase in both Treg proportions and intracellular levels of the lineage-defining transcription factor FoxP3, which correlated with poor outcomes. Accordingly, these Tregs over-expressed a range of suppressive effectors, but also pro-inflammatory molecules like IL32. Most strikingly, they acquired similarity to tumor-infiltrating Tregs, known to suppress local anti-tumor responses. These traits were most marked in acute patients with severe disease, but persisted somewhat in convalescent patients. These results suggest that Tregs may play nefarious roles in COVID-19, via suppressing anti-viral T cell responses during the severe phase of the disease, and/or via a direct pro-inflammatory role.
Collapse
|
13
|
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
|
14
|
Human cytomegalovirus-encoded MicroRNAs: A master regulator of latent infection. INFECTION GENETICS AND EVOLUTION 2020; 78:104119. [DOI: 10.1016/j.meegid.2019.104119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 11/06/2019] [Accepted: 11/14/2019] [Indexed: 01/08/2023]
|
15
|
Fadaei R, Bagheri N, Heidarian E, Nouri A, Hesari Z, Moradi N, Ahmadi A, Ahmadi R. Serum levels of IL-32 in patients with type 2 diabetes mellitus and its relationship with TNF-α and IL-6. Cytokine 2020; 125:154832. [DOI: 10.1016/j.cyto.2019.154832] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/24/2019] [Accepted: 08/24/2019] [Indexed: 10/26/2022]
|
16
|
Paz H, Tsoi J, Kalbasi A, Grasso CS, McBride WH, Schaue D, Butterfield LH, Maurer DM, Ribas A, Graeber TG, Economou JS. Interleukin 32 expression in human melanoma. J Transl Med 2019; 17:113. [PMID: 30953519 PMCID: PMC6449995 DOI: 10.1186/s12967-019-1862-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023] Open
Abstract
Background Various proinflammatory cytokines can be detected within the melanoma tumor microenvironment. Interleukin 32 (IL32) is produced by T cells, NK cells and monocytes/macrophages, but also by a subset of melanoma cells. We sought to better understand the biology of IL32 in human melanoma. Methods We analyzed RNA sequencing data from 53 in-house established human melanoma cell lines and 479 melanoma tumors from The Cancer Genome Atlas dataset. We evaluated global gene expression patterns associated with IL32 expression. We also evaluated the impact of proinflammatory molecules TNFα and IFNγ on IL32 expression and dedifferentiation in melanoma cell lines in vitro. In order to study the transcriptional regulation of IL32 in these cell lines, we cloned up to 10.5 kb of the 5′ upstream region of the human IL32 gene into a luciferase reporter vector. Results A significant proportion of established human melanoma cell lines express IL32, with its expression being highly correlated with a dedifferentiation genetic signature (high AXL/low MITF). Non IL32-expressing differentiated melanoma cell lines exposed to TNFα or IFNγ can be induced to express the three predominant isoforms (α, β, γ) of IL32. Cis-acting elements within this 5′ upstream region of the human IL32 gene appear to govern both induced and constitutive gene expression. In the tumor microenvironment, IL32 expression is highly correlated with genes related to T cell infiltration, and also positively correlates with high AXL/low MITF dedifferentiated gene signature. Conclusions Expression of IL32 in human melanoma can be induced by TNFα or IFNγ and correlates with a treatment-resistant dedifferentiated genetic signature. Constitutive and induced expression are regulated, in part, by cis-acting sequences within the 5′ upstream region. Electronic supplementary material The online version of this article (10.1186/s12967-019-1862-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Helicia Paz
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Jennifer Tsoi
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA, 90095, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Anusha Kalbasi
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA.,Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - Catherine S Grasso
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - William H McBride
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Lisa H Butterfield
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA.,Department of Surgery, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA.,Department of Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Deena M Maurer
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Antoni Ribas
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA, 90095, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - James S Economou
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA. .,Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA. .,Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA, 90095, USA. .,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
17
|
Santinelli L, Statzu M, Pierangeli A, Frasca F, Bressan A, Pinacchio C, Nonne C, Turriziani O, Antonelli G, d'Ettorre G, Scagnolari C. Increased expression of IL-32 correlates with IFN-γ, Th1 and Tc1 in virologically suppressed HIV-1-infected patients. Cytokine 2019; 120:273-281. [PMID: 30910260 DOI: 10.1016/j.cyto.2019.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [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
|
18
|
Kwon OC, Kim S, Hong S, Lee CK, Yoo B, Chang EJ, Kim YG. Role of IL-32 Gamma on Bone Metabolism in Autoimmune Arthritis. Immune Netw 2018; 18:e20. [PMID: 29984038 PMCID: PMC6026691 DOI: 10.4110/in.2018.18.e20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 12/31/2022] Open
Abstract
IL-32 acts as a pro-inflammatory cytokine by inducing the synthesis of inflammatory molecules as well as promoting the morphological changes involved in the transformation of monocytes into osteoclasts (OCs). Evaluation of the functions of IL-32 has mainly focused on its inflammatory properties, such as involvement in the pathogenesis of various autoimmune diseases. Recently, IL-32 was shown to be involved in bone metabolism, in which it promotes the differentiation and activation of OCs and plays a key role in bone resorption in inflammatory conditions. IL-32γ also regulates bone formation in conditions such as ankylosing spondylitis and osteoporosis. In this review, we summarize the results of recent studies on the role of IL-32γ in bone metabolism in inflammatory arthritis.
Collapse
Affiliation(s)
- Oh Chan Kwon
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Soohyun Kim
- Department of Biomedical Science and Technology, Konkuk University, Seoul 05066, Korea
| | - Seokchan Hong
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Chang-Keun Lee
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Bin Yoo
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Eun-Ju Chang
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
19
|
The urinary and serum levels of IL-32 in children with febrile urinary tract infections. Future Sci OA 2017; 3:FSO242. [PMID: 29134126 PMCID: PMC5674269 DOI: 10.4155/fsoa-2017-0076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/10/2017] [Indexed: 12/18/2022] Open
Abstract
Aim We assessed the urinary and serum levels of IL-32 in pediatric patients with acute pyelonephritis (APN) with and without renal scarring. Results We enrolled children aged 2 months to 16 years with APN. Dimercaptosuccinic acid scans and ultrasonography studies were ordered for all patients, and a voiding. A total of 86 children (aged 57 ± 39 months, 74 [86%] female) were enrolled in this study. Group 1 was composed of 19 (16 female) patients, group 2 of 38 (35 female) patients and group 3 of 29 (23 female) patients. There were no significant differences in the serum and absolute urinary levels of IL-32 (UIL-32) between groups, but the urinary IL-32/creatinine ratio (UIL-32/Cr) was significantly higher in children with pyelonephritis than controls.
Collapse
|
20
|
Bhat S, Gardi N, Hake S, Kotian N, Sawant S, Kannan S, Parmar V, Desai S, Dutt A, Joshi NN. Impact of intra-tumoral IL17A and IL32 gene expression on T-cell responses and lymph node status in breast cancer patients. J Cancer Res Clin Oncol 2017; 143:1745-1756. [PMID: 28470472 PMCID: PMC5863950 DOI: 10.1007/s00432-017-2431-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/20/2017] [Indexed: 02/08/2023]
Abstract
PURPOSE Pro-inflammatory cytokines such as Interleukin-17A (IL17A) and Interleukin-32 (IL32), known to enhance natural killer and T cell responses, are also elevated in human malignancies and linked to poor clinical outcomes. To address this paradox, we evaluated relation between IL17A and IL32 expression and other inflammation- and T cell response-associated genes in breast tumors. METHODS TaqMan-based gene expression analysis was carried out in seventy-eight breast tumors. The association between IL17A and IL32 transcript levels and T cell response genes, ER status as well as lymph node status was also examined in breast tumors from TCGA dataset. RESULTS IL17A expression was detected in 32.7% ER-positive and 84.6% ER-negative tumors, with higher expression in the latter group (26.2 vs 7.1-fold, p < 0.01). ER-negative tumors also showed higher expression of IL32 as opposed to ER-positive tumors (8.7 vs 2.5-fold, p < 0.01). Expression of both IL17A and IL32 genes positively correlated with CCL5, GNLY, TBX21, IL21 and IL23 transcript levels (p < 0.01). Amongst ER-positive tumors, higher IL32 expression significantly correlated with lymph node metastases (p < 0.05). Conversely, in ER-negative subtype, high IL17A and IL32 expression was seen in patients with negative lymph node status (p < 0.05). Tumors with high IL32 and IL17A expression showed higher expression of TH1 response genes studied, an observation validated by similar analysis in the TCGA breast tumors (n=1041). Of note, these tumors were characterized by low expression of a potentially immunosuppressive isoform of IL32 (IL32γ). CONCLUSION These results suggest that high expression of both IL17A and IL32 leads to enhancement of T cell responses. Our study, thus, provides basis for the emergence of strong T cell responses in an inflammatory milieu that have been shown to be associated with better prognosis in ER-negative breast cancer.
Collapse
Affiliation(s)
- Shreyas Bhat
- Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Nilesh Gardi
- Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Sujata Hake
- Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Nirupama Kotian
- Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Sharada Sawant
- Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Sadhana Kannan
- Epidemiology and Clinical Trials Unit, Clinical Research Centre, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Vani Parmar
- Department of Surgical Oncology, Tata Memorial Hospital, Parel, Mumbai, Maharashtra, 400012, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Sangeeta Desai
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai, Maharashtra, 400012, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Amit Dutt
- Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Narendra N Joshi
- Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, 410210, India.
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India.
| |
Collapse
|
21
|
Catalán V, Gómez-Ambrosi J, Rodríguez A, Ramírez B, Valentí V, Moncada R, Landecho MF, Silva C, Salvador J, Frühbeck G. Increased Interleukin-32 Levels in Obesity Promote Adipose Tissue Inflammation and Extracellular Matrix Remodeling: Effect of Weight Loss. Diabetes 2016; 65:3636-3648. [PMID: 27630206 DOI: 10.2337/db16-0287] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 09/04/2016] [Indexed: 12/28/2022]
Abstract
Interleukin (IL)-32 is a recently described cytokine involved in the regulation of inflammation. We aimed to explore whether IL-32 could function as an inflammatory and angiogenic factor in human obesity and obesity-associated type 2 diabetes. Samples obtained from 90 subjects were used in the study. Obese patients exhibited higher expression levels of IL-32 in visceral adipose tissue (AT) as well as in subcutaneous AT and peripheral blood mononuclear cells. IL32 was mainly expressed by stromovascular fraction cells, and its expression was significantly enhanced by inflammatory stimuli and hypoxia, whereas no changes were found after the incubation with anti-inflammatory cytokines. The addition of exogenous IL-32 induced the expression of inflammation and extracellular matrix-related genes in human adipocyte cultures, and IL32-silenced adipocytes showed a downregulation of inflammatory genes. Furthermore, adipocyte-conditioned media obtained from obese patients increased IL32 gene expression in human monocyte cultures, whereas the adipocyte-conditioned media from lean volunteers had no effect on IL32 mRNA levels. These findings provide evidence, for the first time, about the inflammatory and remodeling properties of IL-32 in AT, implicating this cytokine in obesity-associated comorbidities.
Collapse
Affiliation(s)
- Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y Nutrición, CIBEROBN, Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y Nutrición, CIBEROBN, Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y Nutrición, CIBEROBN, Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y Nutrición, CIBEROBN, Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
| | - Víctor Valentí
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y Nutrición, CIBEROBN, Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y Nutrición, CIBEROBN, Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
- Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Manuel F Landecho
- Department of Internal Medicine, Clínica Universidad de Navarra, Pamplona, Spain
- TRUEHF Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Camilo Silva
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y Nutrición, CIBEROBN, Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Salvador
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y Nutrición, CIBEROBN, Instituto de Salud Carlos III, Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y Nutrición, CIBEROBN, Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
22
|
Asif AR, Qadri S, Ijaz N, Javed R, Ansari AR, Awais M, Younus M, Riaz H, Du X. Genetic signature of strong recent positive selection at interleukin-32 gene in goat. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2016; 30:912-919. [PMID: 27165029 PMCID: PMC5495668 DOI: 10.5713/ajas.15.0941] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/06/2016] [Accepted: 03/25/2016] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Identification of the candidate genes that play key roles in phenotypic variations can provide new information about evolution and positive selection. Interleukin (IL)-32 is involved in many biological processes, however, its role for the immune response against various diseases in mammals is poorly understood. Therefore, the current investigation was performed for the better understanding of the molecular evolution and the positive selection of single nucleotide polymorphisms in IL-32 gene. METHODS By using fixation index (FST ) based method, IL-32 (9375) gene was found to be outlier and under significant positive selection with the provisional combined allocation of mean heterozygosity and FST . Using nucleotide sequences of 11 mammalian species from National Center for Biotechnology Information database, the evolutionary selection of IL-32 gene was determined using Maximum likelihood model method, through four models (M1a, M2a, M7, and M8) in Codeml program of phylogenetic analysis by maximum liklihood. RESULTS IL-32 is detected under positive selection using the FST simulations method. The phylogenetic tree revealed that goat IL-32 was in close resemblance with sheep IL-32. The coding nucleotide sequences were compared among 11 species and it was found that the goat IL-32 gene shared identity with sheep (96.54%), bison (91.97%), camel (58.39%), cat (56.59%), buffalo (56.50%), human (56.13%), dog (50.97%), horse (54.04%), and rabbit (53.41%) respectively. CONCLUSION This study provides evidence for IL-32 gene as under significant positive selection in goat.
Collapse
Affiliation(s)
- Akhtar Rasool Asif
- Key Lab of Animal Genetics, Breeding and Reproduction of Ministry Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China.,Theriogenology Department, College of Veterinary and Animal Science, Jhang, Sub campus of University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Sumayyah Qadri
- Theriogenology Department, College of Veterinary and Animal Science, Jhang, Sub campus of University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Nabeel Ijaz
- Key Lab of Animal Genetics, Breeding and Reproduction of Ministry Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Ruheena Javed
- Key Lab of Animal Genetics, Breeding and Reproduction of Ministry Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Abdur Rahman Ansari
- Theriogenology Department, College of Veterinary and Animal Science, Jhang, Sub campus of University of Veterinary and Animal Sciences, Lahore 54000, Pakistan.,Department of Anatomy, Histology and Embryology, College of Animal and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Muhammd Awais
- Key Lab of Animal Genetics, Breeding and Reproduction of Ministry Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Muhammad Younus
- Theriogenology Department, College of Veterinary and Animal Science, Jhang, Sub campus of University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Hasan Riaz
- Department of Biosciences, COMSATS Institute of Information Technology, Sahiwal 57000, Pakistan
| | - Xiaoyong Du
- Key Lab of Animal Genetics, Breeding and Reproduction of Ministry Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China.,Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan China
| |
Collapse
|
23
|
Lin HR, Wu YH, Yen WC, Yang CM, Chiu DTY. Diminished COX-2/PGE2-Mediated Antiviral Response Due to Impaired NOX/MAPK Signaling in G6PD-Knockdown Lung Epithelial Cells. PLoS One 2016; 11:e0153462. [PMID: 27097228 PMCID: PMC4838297 DOI: 10.1371/journal.pone.0153462] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/30/2016] [Indexed: 11/18/2022] Open
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) provides the reducing agent NADPH to meet the cellular needs for reductive biosynthesis and the maintenance of redox homeostasis. G6PD-deficient cells experience a high level of oxidative stress and an increased susceptibility to viral infections. Cyclooxygenase-2 (COX-2) is a key mediator in the regulation of viral replication and inflammatory response. In the current study, the role of G6PD on the inflammatory response was determined in both scramble control and G6PD-knockdown (G6PD-kd) A549 cells upon tumor necrosis factor-α (TNF-α) stimulation. A decreased expression pattern of induced COX-2 and reduced production of downstream PGE2 occurred upon TNF-α stimulation in G6PD-kd A549 cells compared with scramble control A549 cells. TNF-α-induced antiviral activity revealed that decreased COX-2 expression enhanced the susceptibility to coronavirus 229E infection in G6PD-kd A549 cells and was a result of the decreased phosphorylation levels of MAPK (p38 and ERK1/2) and NF-κB. The impaired inflammatory response in G6PD-kd A549 cells was found to be mediated through NADPH oxidase (NOX) signaling as elucidated by cell pretreatment with a NOX2-siRNA or NOX inhibitor, diphenyleneiodonium chloride (DPI). In addition, NOX activity with TNF-α treatment in G6PD-kd A549 cells was not up-regulated and was coupled with a decrease in NOX subunit expression at the transcriptional level, implying that TNF-α-mediated NOX signaling requires the participation of G6PD. Together, these data suggest that G6PD deficiency affects the cellular inflammatory response and the decreased TNF-α-mediated antiviral response in G6PD-kd A549 cells is a result of dysregulated NOX/MAPK/NF-κB/COX-2 signaling.
Collapse
Affiliation(s)
- Hsin-Ru Lin
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
| | - Yi-Hsuan Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Wei-Chen Yen
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chuen-Mao Yang
- Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan
- Department of physiology and pharmacology, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- * E-mail: (DTYC); (CMY)
| | - Daniel Tsun-Yee Chiu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan
- Department of Pediatric Hematology, Chang Gung Memorial Hospital, Lin-Kou, Taiwan
- * E-mail: (DTYC); (CMY)
| |
Collapse
|
24
|
IL-32: A Novel Pluripotent Inflammatory Interleukin, towards Gastric Inflammation, Gastric Cancer, and Chronic Rhino Sinusitis. Mediators Inflamm 2016; 2016:8413768. [PMID: 27143819 PMCID: PMC4837279 DOI: 10.1155/2016/8413768] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/23/2016] [Accepted: 03/20/2016] [Indexed: 12/15/2022] Open
Abstract
A vast variety of nonstructural proteins have been studied for their key roles and involvement in a number of biological phenomenona. Interleukin-32 is a novel cytokine whose presence has been confirmed in most of the mammals except rodents. The IL-32 gene was identified on human chromosome 16 p13.3. The gene has eight exons and nine splice variants, namely, IL-32α, IL-32β, IL-32γ, IL-32δ, IL-32ε, IL-32ζ, IL-32η, IL-32θ, and IL-32s. It was found to induce the expression of various inflammatory cytokines including TNF-α, IL-6, and IL-1β as well as macrophage inflammatory protein-2 (MIP-2) and has been reported previously to be involved in the pathogenesis and progression of a number of inflammatory disorders, namely, inflammatory bowel disease (IBD), gastric inflammation and cancer, rheumatoid arthritis, and chronic obstructive pulmonary disease (COPD). In the current review, we have highlighted the involvement of IL-32 in gastric cancer, gastric inflammation, and chronic rhinosinusitis. We have also tried to explore various mechanisms suspected to induce the expression of this extraordinary cytokine as well as various mechanisms of action employed by IL-32 during the mediation and progression of the above said problems.
Collapse
|