1
|
Stafford LS, Plummer CE, Smith WC, Gibson DJ, Sharma J, Vicuna V, Diakite S, Larkin J. A peptide mimic of SOCS1 modulates equine peripheral immune cells in vitro and ocular effector functions in vivo: implications for recurrent uveitis. Front Immunol 2025; 15:1513157. [PMID: 39867889 PMCID: PMC11757128 DOI: 10.3389/fimmu.2024.1513157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/17/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction Recurrent uveitis (RU), an autoimmune disease, is a leading cause of ocular detriment in humans and horses. Equine and human RU share many similarities including spontaneous disease and aberrant cytokine signaling. Reduced levels of SOCS1, a critical regulator of cytokine signaling, is associated with several autoimmune diseases. Topical administration of SOCS1-KIR, a peptide mimic of SOCS1, was previously correlated to reduced ocular pathologies within ERU patients. Methods To further assess the translational potential of a SOCS1 mimetic to treat RU, we assessed peptide-mediated modulation of immune functions in vitro, using equine peripheral blood mononuclear cells (PBMC), and in vivo through topical administration of SOCS1-KIR into the eyes of experimental (non-uveitic) horses. Equine PBMCs from non-uveitic control and ERU horses were cultured with or without SOCS1-KIR pretreatment, followed by 72 hours of mitogen stimulation. Proliferation was assessed using MTT, and cytokine production within cell supernatants was assessed by Luminex. SOCS1-KIR or carrier eye-drops were topically applied to experimental horse eyes twice daily for 21 days, followed by enucleation and isolation of ocular aqueous and vitreous humor. Histology was used to assess peptide treatment safety and localization within treated equine eyes. Cytokine secretion within aqueous humor and vitreous, isolated from experimental equine eyes, was measured by Luminex. Results Following SOCS1-KIR pretreatment, cell proliferation significantly decreased in control, but not ERU-derived PBMCs. Despite differential regulation of cellular proliferation, SOCS1-KIR significantly reduced TNFα and IL-10 secretion in PHA-stimulated control and ERU equine PBMC. SOCS1-KIR increased PBMC secretion of IL-8. Topically administered SOCS1-KIR was well tolerated. Although SOCS1-KIR was undetectable within the eye, topically treated equine eyes had significant reductions in TNFα and IL-10. Interestingly, we found that while SOCS1-KIR treatment reduced TNFα and IL-10 production in healthy and ERU PBMC, SOCS1-KIR differentially modulated proliferation, IP-10 production, and RANTES within these two groups suggesting possible differences in cell types or activation status. Discussion Topical administration of a SOCS1 peptide mimic is safe to the equine eye and reduces ERU associated cytokines IL-10 and TNFα serving as potential biomarkers of drug efficacy in a future clinical trial.
Collapse
Affiliation(s)
- Lauren Stewart Stafford
- Microbiology and Cell Science, Institute of Food and Agricultural Science, University of Florida, Gainesville, FL, United States
| | - Caryn E. Plummer
- Departments of Large and Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - W. Clay Smith
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Daniel J. Gibson
- Capstone College of Nursing, University of Alabama, Tuscaloosa, AL, United States
| | - Jatin Sharma
- Microbiology and Cell Science, Institute of Food and Agricultural Science, University of Florida, Gainesville, FL, United States
| | - Valeria Vicuna
- Microbiology and Cell Science, Institute of Food and Agricultural Science, University of Florida, Gainesville, FL, United States
| | - Sisse Diakite
- Microbiology and Cell Science, Institute of Food and Agricultural Science, University of Florida, Gainesville, FL, United States
| | - Joseph Larkin
- Microbiology and Cell Science, Institute of Food and Agricultural Science, University of Florida, Gainesville, FL, United States
| |
Collapse
|
2
|
Zhou W, Yuan W, Chen Y, Li C, Hu L, Li Q, Wang J, Xue R, Sun Y, Xia Q, Hu L, Wei Y, He M. Single-cell transcriptomics reveals the pulmonary inflammation induced by inhalation of subway fine particles. JOURNAL OF HAZARDOUS MATERIALS 2024; 463:132896. [PMID: 37951166 DOI: 10.1016/j.jhazmat.2023.132896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/28/2023] [Accepted: 10/28/2023] [Indexed: 11/13/2023]
Abstract
People generally take the subway and inevitably inhale the fine particles (PM2.5) on subway platforms. This study revealed whether and how subway PM2.5 causes lung inflammation. Herein, the pulmonary inflammatory response to subway PM2.5 was observed in mice, manifesting as the inflammatory cells infiltration and collagen deposition in tissue, inflammatory cytokine enhancement in bronchoalveolar lavage fluid and Toll-like receptors signal pathway activation in the lungs. Furthermore, single-cell RNA sequencing unearthed subway PM2.5-induced cell-specific responses in the lungs. Twenty immune subsets were identified by the molecular and functional properties. Specific cell populations of CD4+ T and γδ T cells were regarded as the predominant sources of pneumonitis induced by subway PM2.5. Moreover, we demonstrated that the lung inflammatory injury was significantly more attenuated in Rag1-/- mice lacking functional T cells and B cells than that in wild type mice. We proved the slight inflammation of lung tissue in Rag1-/- mice may be dependent on monocytes and neutrophils by activation of the intracellular molecular network. This is the first experimental study on subway PM2.5 causing pulmonary inflammatory damage. It will set an alarm for people who usually travel by subway and efficient measures to reduce PM2.5 should be developed in subway stations.
Collapse
Affiliation(s)
- Weilai Zhou
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Wenke Yuan
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Yuwei Chen
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Chao Li
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, China
| | - Liwen Hu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510275, China
| | - Qidian Li
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Jiawei Wang
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Rou Xue
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Yuan Sun
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Qing Xia
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Longji Hu
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Yuan Wei
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Miao He
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China.
| |
Collapse
|
3
|
Fadanni GP, Calixto JB. Recent progress and prospects for anti-cytokine therapy in preclinical and clinical acute lung injury. Cytokine Growth Factor Rev 2023; 71-72:13-25. [PMID: 37481378 DOI: 10.1016/j.cytogfr.2023.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/10/2023] [Indexed: 07/24/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a heterogeneous cause of respiratory failure that has a rapid onset, a high mortality rate, and for which there is no effective pharmacological treatment. Current evidence supports a critical role of excessive inflammation in ARDS, resulting in several cytokines, cytokine receptors, and proteins within their downstream signalling pathways being putative therapeutic targets. However, unsuccessful trials of anti-inflammatory drugs have thus far hindered progress in the field. In recent years, the prospects of precision medicine and therapeutic targeting of cytokines coevolving into effective treatments have gained notoriety. There is an optimistic and growing understanding of ARDS subphenotypes as well as advances in treatment strategies and clinical trial design. Furthermore, large trials of anti-cytokine drugs in patients with COVID-19 have provided an unprecedented amount of information that could pave the way for therapeutic breakthroughs. While current clinical and nonclinical ARDS research suggest relatively limited potential in monotherapy with anti-cytokine drugs, combination therapy has emerged as an appealing strategy and may provide new perspectives on finding safe and effective treatments. Accurate evaluation of these drugs, however, also relies on well-founded experimental research and the implementation of biomarker-guided stratification in future trials. In this review, we provide an overview of anti-cytokine therapy for acute lung injury and ARDS, highlighting the current preclinical and clinical evidence for targeting the main cytokines individually and the therapeutic prospects for combination therapy.
Collapse
Affiliation(s)
- Guilherme Pasetto Fadanni
- Centre of Innovation and Preclinical Studies (CIEnP), Florianópolis, Santa Catarina, Brazil; Department of Pharmacology, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil.
| | - João Batista Calixto
- Centre of Innovation and Preclinical Studies (CIEnP), Florianópolis, Santa Catarina, Brazil; Department of Pharmacology, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
4
|
Uysalol M, Serin I, Oyacı Y, Yıldız R, Uysalol E, Pehlivan S. Association of Tumor Necrosis Factor-Alpha (TNF-α) and Suppressor of Cytokine Signaling-1 (SOCS-1) Gene Variants in Children with COVID-19. J PEDIAT INF DIS-GER 2023. [DOI: 10.1055/s-0042-1759801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Abstract
Objective The suppressor of cytokine signaling-1 (SOCS-1) gene is an essential physiological regulator of cytokine signaling. Tumor necrosis factor-α (TNF-α) is an important component of the immunological response. Herein, we aimed to investigate the effects of SOCS-1 (-1478 CA > Del) and TNF-α (-308) polymorphisms on disease susceptibility and prognosis in pediatric patients with coronavirus disease 2019 (COVID-19).
Methods One-hundred fifty COVID-19 patients in the COVID-19 emergency department between September 2020 and April 2021 and 80 healthy volunteers (control group) without any additional disease were included. Baseline gene polymorphisms were compared between the patient and healthy control groups. Afterward, the gene polymorphism distribution was examined by forming two separate clinical patients' subgroups.
Results While CA/CA and CA/Del gene variants of SOCS-1 were higher in the patient group, Del/Del genotype was more common in the control group (p < 0.05). The GG genotype of the TNF-α was significantly more common in the severe subgroup (p = 0.044). The GA genotype of TNF-α was associated with the risk of hospitalization (2.83-fold), while the GG genotype was found to be protective in terms of hospitalization (2.95-fold).
Conclusions This study will be a guide in terms of the presence of high cytokine release genotypes and COVID-19-related cytokine release syndromes.
Collapse
Affiliation(s)
- Metin Uysalol
- Department of Pediatrics, Division of Pediatric Emergency, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| | - Istemi Serin
- Department of Hematology, Istanbul Training and Research Hospital, University of Health Science, Istanbul, Türkiye
| | - Yasemin Oyacı
- Department of Medical Biology and Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| | - Raif Yıldız
- Department of Pediatrics, Division of Pediatric Emergency, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| | - Ezgi Uysalol
- Department of Pediatric Hematology- Oncology, Basaksehir Cam and Sakura City Hospital, Istanbul, Türkiye
| | - Sacide Pehlivan
- Department of Medical Biology and Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| |
Collapse
|
5
|
Bhattacharjee R, Das SS, Biswal SS, Nath A, Das D, Basu A, Malik S, Kumar L, Kar S, Singh SK, Upadhye VJ, Iqbal D, Almojam S, Roychoudhury S, Ojha S, Ruokolainen J, Jha NK, Kesari KK. Mechanistic Role of HPV-Associated Early Proteins in Cervical Cancer: Molecular Pathways and Targeted Therapeutic Strategies. Crit Rev Oncol Hematol 2022; 174:103675. [DOI: 10.1016/j.critrevonc.2022.103675] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 12/24/2022] Open
|
6
|
Verma AK, Bauer C, Palani S, Metzger DW, Sun K. IFN-γ Drives TNF-α Hyperproduction and Lethal Lung Inflammation during Antibiotic Treatment of Postinfluenza Staphylococcus aureus Pneumonia. THE JOURNAL OF IMMUNOLOGY 2021; 207:1371-1376. [PMID: 34380647 DOI: 10.4049/jimmunol.2100328] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/29/2021] [Indexed: 01/15/2023]
Abstract
Inflammatory cytokine storm is a known cause for acute respiratory distress syndrome. In this study, we have investigated the role of IFN-γ in lethal lung inflammation using a mouse model of postinfluenza methicillin-resistant Staphylococcus aureus (MRSA) pneumonia. To mimic the clinical scenario, animals were treated with antibiotics for effective bacterial control following MRSA superinfection. However, antibiotic therapy alone is not sufficient to improve survival of wild-type animals in this lethal acute respiratory distress syndrome model. In contrast, antibiotics induce effective protection in mice deficient in IFN-γ response. Mechanistically, we show that rather than inhibiting bacterial clearance, IFN-γ promotes proinflammatory cytokine response to cause lethal lung damage. Neutralization of IFN-γ after influenza prevents hyperproduction of TNF-α, and thereby protects against inflammatory lung damage and animal mortality. Taken together, the current study demonstrates that influenza-induced IFN-γ drives a stepwise propagation of inflammatory cytokine response, which ultimately results in fatal lung damage during secondary MRSA pneumonia, despite of antibiotic therapy.
Collapse
Affiliation(s)
- Atul K Verma
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Christopher Bauer
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Sunil Palani
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX; and
| | - Dennis W Metzger
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY
| | - Keer Sun
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE; .,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX; and
| |
Collapse
|
7
|
Sood N, Verma DK, Paria A, Yadav SC, Yadav MK, Bedekar MK, Kumar S, Swaminathan TR, Mohan CV, Rajendran KV, Pradhan PK. Transcriptome analysis of liver elucidates key immune-related pathways in Nile tilapia Oreochromis niloticus following infection with tilapia lake virus. FISH & SHELLFISH IMMUNOLOGY 2021; 111:208-219. [PMID: 33577877 DOI: 10.1016/j.fsi.2021.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 06/12/2023]
Abstract
Nile tilapia (Oreochromis niloticus) is one of the most important aquaculture species farmed worldwide. However, the recent emergence of tilapia lake virus (TiLV) disease, also known as syncytial hepatitis of tilapia, has threatened the global tilapia industry. To gain more insight regarding the host response against the disease, the transcriptional profiles of liver in experimentally-infected and control tilapia were compared. Analysis of RNA-Seq data identified 4640 differentially expressed genes (DEGs), which were involved among others in antigen processing and presentation, MAPK, apoptosis, necroptosis, chemokine signaling, interferon, NF-kB, acute phase response and JAK-STAT pathways. Enhanced expression of most of the DEGs in the above pathways suggests an attempt by tilapia to resist TiLV infection. However, upregulation of some of the key genes such as BCL2L1 in apoptosis pathway; NFKBIA in NF-kB pathway; TRFC in acute phase response; and SOCS, EPOR, PI3K and AKT in JAK-STAT pathway and downregulation of the genes, namely MAP3K7 in MAPK pathway; IFIT1 in interferon; and TRIM25 in NF-kB pathway suggested that TiLV was able to subvert the host immune response to successfully establish the infection. The study offers novel insights into the cellular functions that are affected following TiLV infection and will serve as a valuable genomic resource towards our understanding of susceptibility of tilapia to TiLV infection.
Collapse
Affiliation(s)
- Neeraj Sood
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Dev Kumar Verma
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Anutosh Paria
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Shrish Chandra Yadav
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Manoj Kumar Yadav
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Megha Kadam Bedekar
- ICAR-Central Institute of Fisheries Education, Versova, Andheri (W), Mumbai, 400 061, Maharashtra, India
| | - Saurav Kumar
- ICAR-Central Institute of Fisheries Education, Versova, Andheri (W), Mumbai, 400 061, Maharashtra, India
| | - Thangaraj Raja Swaminathan
- Peninsular and Marine Fish Genetic Resources Centre, ICAR-NBFGR, CMFRI Campus, Kochi, 682 018, Kerala, India
| | | | - K V Rajendran
- ICAR-Central Institute of Fisheries Education, Versova, Andheri (W), Mumbai, 400 061, Maharashtra, India
| | - Pravata Kumar Pradhan
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India.
| |
Collapse
|
8
|
Oh SJ, Gim JA, Lee JK, Park H, Shin OS. Coxsackievirus B3 Infection of Human Neural Progenitor Cells Results in Distinct Expression Patterns of Innate Immune Genes. Viruses 2020; 12:v12030325. [PMID: 32192194 PMCID: PMC7150933 DOI: 10.3390/v12030325] [Citation(s) in RCA: 10] [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: 02/13/2020] [Revised: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022] Open
Abstract
Coxsackievirus B3 (CVB3), a member of Picornaviridae family, is an important human pathogen that causes a wide range of diseases, including myocarditis, pancreatitis, and meningitis. Although CVB3 has been well demonstrated to target murine neural progenitor cells (NPCs), gene expression profiles of CVB3-infected human NPCs (hNPCs) has not been fully explored. To characterize the molecular signatures and complexity of CVB3-mediated host cellular responses in hNPCs, we performed QuantSeq 3′ mRNA sequencing. Increased expression levels of viral RNA sensors (RIG-I, MDA5) and interferon-stimulated genes, such as IFN-β, IP-10, ISG15, OAS1, OAS2, Mx2, were detected in response to CVB3 infection, while IFN-γ expression level was significantly downregulated in hNPCs. Consistent with the gene expression profile, CVB3 infection led to enhanced secretion of inflammatory cytokines and chemokines, such as interleukin-6 (IL-6), interleukin-8 (IL-8), and monocyte chemoattractant protein-1 (MCP-1). Furthermore, we show that type I interferon (IFN) treatment in hNPCs leads to significant attenuation of CVB3 RNA copy numbers, whereas, type II IFN (IFN-γ) treatment enhances CVB3 replication and upregulates suppressor of cytokine signaling 1/3 (SOCS) expression levels. Taken together, our results demonstrate the distinct molecular patterns of cellular responses to CVB3 infection in hNPCs and the pro-viral function of IFN-γ via the modulation of SOCS expression.
Collapse
Affiliation(s)
- Soo-Jin Oh
- Department of Biomedical Sciences, BK21 PLUS program, College of Medicine, Korea University Guro Hospital, Seoul 08308, Korea; (S.-J.O.); (J.K.L.)
| | - Jeong-An Gim
- Medical Science Research Center, College of Medicine, Korea University Guro Hospital, Seoul 08308, Korea;
| | - Jae Kyung Lee
- Department of Biomedical Sciences, BK21 PLUS program, College of Medicine, Korea University Guro Hospital, Seoul 08308, Korea; (S.-J.O.); (J.K.L.)
| | - Hosun Park
- Department of Microbiology, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Namgu, Daegu 42415, Korea
- Correspondence: (H.P.); (O.S.S.); Tel.: +82-53-640-6943 (H.P.); +82-2-2626-3280 (O.S.S.)
| | - Ok Sarah Shin
- Department of Biomedical Sciences, BK21 PLUS program, College of Medicine, Korea University Guro Hospital, Seoul 08308, Korea; (S.-J.O.); (J.K.L.)
- Correspondence: (H.P.); (O.S.S.); Tel.: +82-53-640-6943 (H.P.); +82-2-2626-3280 (O.S.S.)
| |
Collapse
|
9
|
Zika Virus-Induction of the Suppressor of Cytokine Signaling 1/3 Contributes to the Modulation of Viral Replication. Pathogens 2020; 9:pathogens9030163. [PMID: 32120897 PMCID: PMC7157194 DOI: 10.3390/pathogens9030163] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/15/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that has emerged and caused global outbreaks since 2007. Although ZIKV proteins have been shown to suppress early anti-viral innate immune responses, little is known about the exact mechanisms. This study demonstrates that infection with either the African or Asian lineage of ZIKV leads to a modulated expression of suppressor of cytokine signaling (SOCS) genes encoding SOCS1 and SOCS3 in the following cell models: A549 human lung adenocarcinoma cells; JAr human choriocarcinoma cells; human neural progenitor cells. Studies of viral gene expression in response to SOCS1 or SOCS3 demonstrated that the knockdown of these SOCS proteins inhibited viral NS5 or ZIKV RNA expression, whereas overexpression resulted in an increased expression. Moreover, the overexpression of SOCS1 or SOCS3 inhibited the retinoic acid-inducible gene-I-like receptor-mediated activation of both type I and III interferon pathways. These results imply that SOCS upregulation following ZIKV infection modulates viral replication, possibly via the regulation of anti-viral innate immune responses.
Collapse
|
10
|
Pal A, Kundu R. Human Papillomavirus E6 and E7: The Cervical Cancer Hallmarks and Targets for Therapy. Front Microbiol 2020; 10:3116. [PMID: 32038557 PMCID: PMC6985034 DOI: 10.3389/fmicb.2019.03116] [Citation(s) in RCA: 333] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 12/24/2019] [Indexed: 01/14/2023] Open
Abstract
Human papillomavirus (HPV)-induced cervical cancer is a major health issue among women from the poorly/under-developed sectors of the world. It accounts for a high-mortality rate because of its late diagnosis and poor prognosis. Initial establishment and subsequent progression of this form of cancer are completely dependent on two major oncogenes E6 and E7, which are expressed constitutively leading to tumorigenesis. Thus, manipulation of these genes represents the most successful form of cervical cancer therapy. In the present article, information on structural, functional, and clinical dimensions of E6 and E7 activity has been reviewed. The genome organization and protein structure of E6 and E7 have been discussed followed by their mechanism to establish the six major cancer hallmarks in cervical tissues for tumor propagation. The later section of this review article deals with the different modes of therapeutics, which functions by deregulating E6 and E7 activity. Since E6 and E7 are the biomarkers of a cervical cancer cell and are the ones driving the cancer progression, therapeutic approaches targeting E6 and E7 have been proved to be highly efficient in terms of focused removal of abnormally propagating malignant cells. Therapeutics including different forms of vaccines to advanced genome editing techniques, which suppress E6 and E7 activity, have been found to successfully bring down the population of cervical cancer cells infected with HPV. T-cell mediated immunotherapy is another upcoming successful form of treatment to eradicate HPV-infected tumorigenic cells. Additionally, therapeutics using natural compounds from plants or other natural repositories, i.e., phytotherapeutic approaches have also been reviewed here, which prove their anticancer potential through E6 and E7 inhibitory effects. Thus, E6 and E7 repression through any of these methods is a significant approach toward cervical cancer therapy, described in details in this review along with an insight into the signaling pathways and molecular mechanistic of E6 and E7 action.
Collapse
Affiliation(s)
| | - Rita Kundu
- Cell Biology Laboratory, Department of Botany, Centre of Advanced Studies, University of Calcutta, Kolkata, India
| |
Collapse
|
11
|
LeMessurier KS, Iverson AR, Chang TC, Palipane M, Vogel P, Rosch JW, Samarasinghe AE. Allergic inflammation alters the lung microbiome and hinders synergistic co-infection with H1N1 influenza virus and Streptococcus pneumoniae in C57BL/6 mice. Sci Rep 2019; 9:19360. [PMID: 31852944 PMCID: PMC6920369 DOI: 10.1038/s41598-019-55712-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/28/2019] [Indexed: 12/12/2022] Open
Abstract
Asthma is a chronic airways condition that can be exacerbated during respiratory infections. Our previous work, together with epidemiologic findings that asthmatics were less likely to suffer from severe influenza during the 2009 pandemic, suggest that additional complications of influenza such as increased susceptibility to bacterial superinfection, may be mitigated in allergic hosts. To test this hypothesis, we developed a murine model of 'triple-disease' in which mice rendered allergic to Aspergillus fumigatus were co-infected with influenza A virus and Streptococcus pneumoniae seven days apart. Significant alterations to known synergistic effects of co-infection were noted in the allergic mice including reduced morbidity and mortality, bacterial burden, maintenance of alveolar macrophages, and reduced lung inflammation and damage. The lung microbiome of allergic mice differed from that of non-allergic mice during co-infection and antibiotic-induced perturbation to the microbiome rendered allergic animals susceptible to severe morbidity. Our data suggest that responses to co-infection in allergic hosts likely depends on the immune and microbiome states and that antibiotics should be used with caution in individuals with underlying chronic lung disease.
Collapse
Affiliation(s)
- Kim S LeMessurier
- Department of Paediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA
- Children's Foundation Research Institute, Memphis, TN, 38103, USA
| | - Amy R Iverson
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Maneesha Palipane
- Department of Paediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA
- Children's Foundation Research Institute, Memphis, TN, 38103, USA
| | - Peter Vogel
- Department of Veterinary Pathology at St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jason W Rosch
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Amali E Samarasinghe
- Department of Paediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA.
- Children's Foundation Research Institute, Memphis, TN, 38103, USA.
| |
Collapse
|
12
|
Liu S, Yan R, Chen B, Pan Q, Chen Y, Hong J, Zhang L, Liu W, Wang S, Chen JL. Influenza Virus-Induced Robust Expression of SOCS3 Contributes to Excessive Production of IL-6. Front Immunol 2019; 10:1843. [PMID: 31474976 PMCID: PMC6706793 DOI: 10.3389/fimmu.2019.01843] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Influenza A virus (IAV) remains a major public health threat in the world, as indicated by the severe pneumonia caused by its infection annually. Interleukin-6 (IL-6) involved excessive inflammatory response to IAV infection profoundly contributes to the virus pathogenesis. However, the precise mechanisms underlying such a response are poorly understood. Here we found from both in vivo and in vitro studies that IAV not only induced a surge of IL-6 release, but also greatly upregulated expression of suppressor of cytokine signaling-3 (SOCS3), the potent suppressor of IL-6-associated signal transducer and activator of transcription 3 (STAT3) signaling. Interestingly, there existed a cytokine-independent mechanism of the robust induction of SOCS3 by IAV at early stages of the infection. Furthermore, we employed SOCS3-knockdown transgenic mice (TG), and surprisingly observed from virus challenge experiments using these mice that disruption of SOCS3 expression provided significant protection against IAV infection, as evidenced by attenuated acute lung injury, a higher survival rate of infected animals and lower viral load in infected tissues as compared with those of wild-type littermates under the same condition. The activity of nuclear factor-kappa B (NFκB) and the expression of its target gene IL-6 were suppressed in SOCS3-knockdown A549 cells and the TG mice after infection with IAV. Moreover, we defined that enhanced STAT3 activity caused by SOCS3 silencing was important for the regulation of NFκB and IL-6. These findings establish a critical role for IL-6-STAT3-SOCS3 axis in the pathogenesis of IAV and suggest that influenza virus may have evolved a strategy to circumvent IL-6/STAT3-mediated immune response through upregulating SOCS3.
Collapse
Affiliation(s)
- Shasha Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ruoxiang Yan
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Biao Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qidong Pan
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yuhai Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jinxuan Hong
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Lianfeng Zhang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Beijing, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Song Wang
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ji-Long Chen
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
13
|
SOCS-1 Suppresses Inflammation Through Inhibition of NALP3 Inflammasome Formation in Smoke Inhalation-Induced Acute Lung Injury. Inflammation 2019; 41:1557-1567. [PMID: 29907905 PMCID: PMC7102050 DOI: 10.1007/s10753-018-0802-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Smoke inhalation leads to acute lung injury (ALI), a devastating clinical problem associated with high mortality rates. Suppressor of cytokine signaling-1 (SOCS-1) is a negative regulator of proinflammatory cytokine signaling. We have found that adenoviral gene transfer of SOCS-1 ameliorates smoke inhalation-induced lung injury in C57BL/6 mice. We also found that the release of adenosine triphosphate (ATP) was increased post smoke exposure, while oxidized ATP, an inhibitor of purinergic P2X7 receptor, suppressed smoke-induced NALP3 inflammasome assembly, caspase-1 activation, and K+ efflux. Similar to oxidized ATP, high protein level of SOCS-1 dampened the formation of NALP3 inflammasome and the activation of caspase-1 and IL-1β induced by smoke exposure in mouse alveolar macrophages. In conclusion, SOCS-1 relieves smoke inhalation-induced pulmonary inflammation and injury by inhibiting NALP3 inflammasome formation.
Collapse
|
14
|
Morgan DJ, Casulli J, Chew C, Connolly E, Lui S, Brand OJ, Rahman R, Jagger C, Hussell T. Innate Immune Cell Suppression and the Link With Secondary Lung Bacterial Pneumonia. Front Immunol 2018; 9:2943. [PMID: 30619303 PMCID: PMC6302086 DOI: 10.3389/fimmu.2018.02943] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
Secondary infections arise as a consequence of previous or concurrent conditions and occur in the community or in the hospital setting. The events allowing secondary infections to gain a foothold have been studied for many years and include poor nutrition, anxiety, mental health issues, underlying chronic diseases, resolution of acute inflammation, primary immune deficiencies, and immune suppression by infection or medication. Children, the elderly and the ill are particularly susceptible. This review is concerned with secondary bacterial infections of the lung that occur following viral infection. Using influenza virus infection as an example, with comparisons to rhinovirus and respiratory syncytial virus infection, we will update and review defective bacterial innate immunity and also highlight areas for potential new investigation. It is currently estimated that one in 16 National Health Service (NHS) hospital patients develop an infection, the most common being pneumonia, lower respiratory tract infections, urinary tract infections and infection of surgical sites. The continued drive to understand the mechanisms of why secondary infections arise is therefore of key importance.
Collapse
Affiliation(s)
- David J Morgan
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Joshua Casulli
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Christine Chew
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Emma Connolly
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Sylvia Lui
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Oliver J Brand
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Rizwana Rahman
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Christopher Jagger
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Tracy Hussell
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
15
|
Xin-Jia-Xiang-Ru-Yin alleviated H1N1-induced acute lung injury and inhibited the IFN-γ-related regulatory pathway in summer flu. Biomed Pharmacother 2018; 108:201-207. [DOI: 10.1016/j.biopha.2018.09.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/01/2018] [Accepted: 09/04/2018] [Indexed: 01/22/2023] Open
|
16
|
Primary Role of Suppressor of Cytokine Signaling 1 in Mycobacterium bovis BCG Infection. Infect Immun 2018; 86:IAI.00376-18. [PMID: 30181351 DOI: 10.1128/iai.00376-18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/29/2018] [Indexed: 11/20/2022] Open
Abstract
Suppressor of cytokine signaling 1 (SOCS1) is a negative regulator of JAK/STAT signaling and is induced by mycobacterial infection. To understand the major function of SOCS1 during infection, we established a novel system in which recombinant Mycobacterium bovis bacillus Calmette-Guérin expressed dominant-negative SOCS1 (rBCG-SOCS1DN) because it would not affect the function of SOCS1 in uninfected cells. When C57BL/6 mice and RAG1-/- mice were intratracheally inoculated with rBCG-SOCS1DN, the amount of rBCG-SOCS1DN in the lungs was significantly reduced compared to that in the lungs of mice inoculated with a vector control counterpart and wild-type BCG. However, these significant differences were not observed in NOS2-/- mice and RAG1-/- NOS2-/- double-knockout mice. These findings demonstrated that SOCS1 inhibits nitric oxide (NO) production to establish mycobacterial infection and that rBCG-SOCS1DN has the potential to be a powerful tool for studying the primary function of SOCS1 in mycobacterial infection.
Collapse
|
17
|
Klonoski JM, Watson T, Bickett TE, Svendsen JM, Gau TJ, Britt A, Nelson JT, Schlenker EH, Chaussee MS, Rynda-Apple A, Huber VC. Contributions of Influenza Virus Hemagglutinin and Host Immune Responses Toward the Severity of Influenza Virus: Streptococcus pyogenes Superinfections. Viral Immunol 2018; 31:457-469. [PMID: 29870311 PMCID: PMC6043403 DOI: 10.1089/vim.2017.0193] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Influenza virus infections can be complicated by bacterial superinfections, which are medically relevant because of a complex interaction between the host, the virus, and the bacteria. Studies to date have implicated several influenza virus genes, varied host immune responses, and bacterial virulence factors, however, the host-pathogen interactions that predict survival versus lethal outcomes remain undefined. Previous work by our group showed that certain influenza viruses could yield a survival phenotype (A/swine/Texas/4199-2/98-H3N2, TX98), whereas others were associated with a lethal phenotype (A/Puerto Rico/8/34-H1N1, PR8). Based on this observation, we developed the hypothesis that individual influenza virus genes could contribute to a superinfection, and that the host response after influenza virus infection could influence superinfection severity. The present study analyzes individual influenza virus gene contributions to superinfection severity using reassortant viruses created using TX98 and PR8 viral genes. Host and pathogen interactions, relevant to survival and lethal phenotypes, were studied with a focus on pathogen clearance, host cellular infiltrates, and cytokine levels after infection. Specifically, we found that the hemagglutinin gene expressed by an influenza virus can contribute to the severity of a secondary bacterial infection, likely through modulation of host proinflammatory responses. Altogether, these results advance our understanding of molecular mechanisms underlying influenza virus-bacteria superinfections and identify viral and corresponding host factors that may contribute to morbidity and mortality.
Collapse
Affiliation(s)
- Joshua M. Klonoski
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Trevor Watson
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Thomas E. Bickett
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Joshua M. Svendsen
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Tonia J. Gau
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Alexandra Britt
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Jeff T. Nelson
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Evelyn H. Schlenker
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Michael S. Chaussee
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Agnieszka Rynda-Apple
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Victor C. Huber
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| |
Collapse
|
18
|
Dumpati R, Ramatenki V, Vadija R, Vellanki S, Vuruputuri U. Structural insights into suppressor of cytokine signaling 1 protein- identification of new leads for type 2 diabetes mellitus. J Mol Recognit 2018; 31:e2706. [PMID: 29630758 DOI: 10.1002/jmr.2706] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/22/2017] [Accepted: 02/04/2018] [Indexed: 12/23/2022]
Abstract
The study considers the Suppressor of cytokine signaling 1 (SOCS1) protein as a novel Type 2 diabetes mellitus (T2DM) drug target. T2DM in human beings is also triggered by the over expression of SOCS proteins. The SOCS1 acts as a ubiquitin ligase (E3), degrades Insulin Receptor Substrate 1 and 2 (IRS1 and IRS2) proteins, and causes insulin resistance. Therefore, the structure of the SOCS1 protein was evaluated using homology-modeling and molecular dynamics methods and validated using standard computational protocols. The Protein-Protein docking study of SOCS1 with its natural substrates, IRS1 and IRS2, and subsequent solvent accessible surface area analysis gave insight into the binding region of the SOCS1 protein. The in silico active site prediction tools highlight the residues Val155 to Ile211 in SOCS1 being implicated in the ubiquitin mediated protein degradation of the proteins IRS1 and IRS2. Virtual screening in the active site region, using large structural databases, results in selective lead structures with 3-Pyridinol, Xanthine, and Alanine moieties as Pharmacophore. The virtual screening study shows that the residues Glu149, Gly187, Arg188, Leu191, and Ser205 of the SOCS1 are important for binding. The docking study with current anti-diabetic therapeutics shows that the drugs Glibenclamide and Glyclopyramide have a partial affinity towards SOCS1. The predicted ADMET and IC50 properties for the identified ligands are within the acceptable range with drug-like properties. The structural data of SOCS1, its active site, and the identified lead structures are expedient in the development of new T2DM therapeutics.
Collapse
Affiliation(s)
- Ramakrishna Dumpati
- Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana State, India
| | - Vishwanath Ramatenki
- Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana State, India
| | - Rajender Vadija
- Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana State, India
| | - Santhiprada Vellanki
- Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana State, India
| | - Uma Vuruputuri
- Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana State, India
| |
Collapse
|
19
|
Zhang W, Chen S, Zhang J, Wu Z, Wang M, Jia R, Zhu D, Liu M, Sun K, Yang Q, Wu Y, Chen X, Cheng A. Molecular identification and immunological characteristics of goose suppressor of cytokine signaling 1 (SOCS-1) in vitro and vivo following DTMUV challenge. Cytokine 2017; 93:1-9. [PMID: 28416080 DOI: 10.1016/j.cyto.2017.03.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/18/2017] [Accepted: 03/31/2017] [Indexed: 12/31/2022]
Abstract
Purpose suppressor of cytokine signaling 1 (SOCS-1) is inducible feedback inhibitors of cytokine signaling and involved in viral infection through regulation of both innate and adaptive immunity. In this study, we firstly cloned SOCS-1 (goSOCS-1) from duck Tembusu virus (DTMUV) infected goose. The full-length sequence of goSOCS-1 ORF is 624bp and encoded 108 amino acids. Structurally, the mainly functional regions (KIR, SH2, SOCS-box) were conserved between avian and mammalian. The tissues distribution data showed SOCS-1 highly expressed in immune related tissues (SP, LU, HG) of both gosling and adult goose. Moreover, the goSOCS-1 transcripts were induced by goIFNs in GEFs and by TLR ligands in PBMCs. Notably, upon DTMUV infection, highly expression level of goSOCS-1 was detected in vitro and in vivo with high viral load. Our results indicated that goSOCS-1 might involve in both innate and adaptive antiviral immunity of waterfowl.
Collapse
Affiliation(s)
- Wei Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| | - Jingyue Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Zhen Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Renyong Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Dekang Zhu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Mafeng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Kunfeng Sun
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Qiao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Ying Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xiaoyue Chen
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| |
Collapse
|
20
|
Zhu X, Bai J, Liu P, Wang X, Jiang P. Suppressor of cytokine signaling 3 plays an important role in porcine circovirus type 2 subclinical infection by downregulating proinflammatory responses. Sci Rep 2016; 6:32538. [PMID: 27581515 PMCID: PMC5007517 DOI: 10.1038/srep32538] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 08/04/2016] [Indexed: 12/12/2022] Open
Abstract
Porcine circovirus type 2 (PCV2) causes porcine circovirus-associated diseases and usually evokes a subclinical infection, without any obvious symptoms, in pigs. It remains unclear how PCV2 leads to a subclinical infection. In this study, we found that peripheral blood mononuclear cells (PBMCs) from PCV2-challenged piglets with no significant clinical symptoms exhibited increased expression of suppressor of cytokine signaling (SOCS) 3, but no significant changes in the expression of the proinflammatory cytokines interleukin (IL)-6 and tumor necrosis factor (TNF)-α; this differed from piglets that displayed significant clinical symptoms. IL-6- and TNF-α-mediated signalings were inhibited in PBMCs from subclinical piglets. Elevated SOCS3 levels inhibited IL-6- and TNF-α-mediated NF-kappa-B inhibitor alpha degradation in PBMCs and PK-15 cells. SOCS3 production was also increased in PCV2-infected PK-15 porcine kidney cells, and IL-6 and TNF-α production that was induced by PCV2 in PK-15 cells was significantly increased when SOCS3 was silenced by a small interfering RNA. SOCS3 interacted with signal transducer and activator of transcription 3 and TNF-associated receptor-associated factor 2, suggesting mechanisms by which SOCS3 inhibits IL-6 and TNF-α signaling. We conclude that SOCS3 plays an important role in PCV2 subclinical infection by suppressing inflammatory responses in primary immune cells.
Collapse
Affiliation(s)
- Xuejiao Zhu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Juan Bai
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Panrao Liu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xianwei Wang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ping Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
21
|
Schneider DJ, Speth JM, Peters-Golden M. Signed, Sealed, Delivered: Microenvironmental Modulation of Extracellular Vesicle-Dependent Immunoregulation in the Lung. Front Cell Dev Biol 2016; 4:94. [PMID: 27626032 PMCID: PMC5004409 DOI: 10.3389/fcell.2016.00094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/18/2016] [Indexed: 12/26/2022] Open
Abstract
Unconventional secretion and subsequent uptake of molecular cargo via extracellular vesicles (EVs) is an important mechanism by which cells can exert paracrine effects. While this phenomenon has been widely characterized in the context of their ability to promote inflammation, less is known about the ability of EVs to transfer immunosuppressive cargo. Maintenance of normal physiology in the lung requires suppression of potentially damaging inflammatory responses to the myriad of insults to which it is continually exposed. Recently, our laboratory has reported the ability of alveolar macrophages (AMs) to secrete suppressors of cytokine signaling (SOCS) proteins within microvesicles (MVs) and exosomes (Exos). Uptake of these EVs by alveolar epithelial cells (AECs) resulted in inhibition of pro-inflammatory STAT activation in response to cytokines. Moreover, AM packaging of SOCS within EVs could be rapidly tuned in response to exogenous or AEC-derived substances. In this article we will highlight gaps in knowledge regarding microenvironmental modulation of cargo packaging and utilization as well as EV secretion and uptake. Advances in these areas are critical for improving understanding of intercellular communication in the immune system and for therapeutic application of artificial vesicles aimed at treatment of diseases characterized by dysregulated inflammation.
Collapse
Affiliation(s)
- Daniel J Schneider
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School USA
| | - Jennifer M Speth
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical SchoolUSA; Graduate Program in Immunology, University of Michigan Medical SchoolUSA
| |
Collapse
|
22
|
Basile K, Dwyer DE, Kok J. Fat and flu: fact or fiction? Future Virol 2016. [DOI: 10.2217/fvl-2016-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Novel risk factors for severe influenza infection were described during the early phases of the influenza pandemic of 2009. Worldwide, the rate of severe influenza in the obese and morbidly obese population was disproportionate to that of the general population. This risk factor has now been recognized presumably due to the increasing prevalence of obesity. The cause behind this trend may extend beyond the known deleterious effects of obesity on respiratory physiology, as emerging evidence in animal models demonstrate A(H1N1)pdm09 itself confers worse outcomes compared with seasonal influenza subtypes. Currently, uncertainty remains regarding the optimal antiviral regimen and vaccination strategies in obese individuals. Therefore, further studies on the effects of obesity on influenza infection need to be prioritized.
Collapse
Affiliation(s)
- Kerri Basile
- Centre for Infectious Diseases & Microbiology Laboratory Services, Institute of Clinical Pathology & Medical Research, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
- Marie Bashir Institute for Infectious Diseases & Biosecurity, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
- Centre for Research Excellence in Critical Infections, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
| | - Dominic E Dwyer
- Centre for Infectious Diseases & Microbiology Laboratory Services, Institute of Clinical Pathology & Medical Research, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
- Marie Bashir Institute for Infectious Diseases & Biosecurity, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
- Centre for Research Excellence in Critical Infections, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
| | - Jen Kok
- Centre for Infectious Diseases & Microbiology Laboratory Services, Institute of Clinical Pathology & Medical Research, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
- Marie Bashir Institute for Infectious Diseases & Biosecurity, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
- Centre for Research Excellence in Critical Infections, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
| |
Collapse
|