201
|
Sun H, Li Y, Quan X, Chen N, Jin X, Jin W, Jin Y, Shen X. PIAS3/SOCS1-STAT3 axis responses to oxidative stress in hepatocellular cancer cells. Am J Transl Res 2021; 13:12395-12409. [PMID: 34956461 PMCID: PMC8661178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/23/2021] [Indexed: 06/14/2023]
Abstract
The participation of STAT3 and its upstream inhibitors, PIAS3 and SOCS1, in the oxidative response of hepatocellular carcinoma (HCC) cells was uncertain. Here, the expression of PIAS3 and SOCS1 in HCC tissues and cell lines was explored, and we sought to determine whether oxidative stress epigenetically regulated PIAS3 and SOCS1 expression and STAT3 activation in HCC cells. The expression of PIAS3 and SOCS1 was markedly decreased in HCC cell lines and tissues compared to normal hepatic cells and tissues. In HCC patients, low PIAS3 and SOCS1 expression were associated with poor survival. Oxidative stress induced by H2O2 in HepG2 cells was indicated by low antioxidant levels and high protein carbonyl content. Moreover, oxidative stress in HepG2 cells contributed to reduced proliferation but increased apoptosis, migration, and invasion capacity, which might be counteracted by antioxidants, such as tocopheryl acetate (TA). PIAS3 and SOCS1 expression was markedly decreased, while STAT3 was activated in HepG2 cells in response to H2O2 exposure. Co-treatment with antioxidant TA effectively increased the expression of PIAS3 and SOCS1, but it dephosphorylated STAT3 in H2O2-treated cells. PIAS1 or SOCS1 overexpression in HepG2 cells after H2O2 treatment restored cell viability and anti-oxidative responses and decreased apoptosis, migration, and invasion ability, and dephosphorylated STAT3 levels. Co-administration of the STAT3 activator, colivelin, partially abolished the effect of PIAS3 and SOCS1 overexpression in these processes. Therefore, oxidative stress in HCC cells may improve their migration and reduce proliferation through STAT3 activation through the repression of PIAS3 and SOCS1 expression.
Collapse
Affiliation(s)
- Honghua Sun
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Yanglong Li
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Xianglan Quan
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Ning Chen
- Department of Infection Disease, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Xinglin Jin
- Department of General Surgert, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Wenbiao Jin
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Yongmin Jin
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Xionghu Shen
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| |
Collapse
|
202
|
Subramaniam S, Anandha Rao JS, Ramdas P, Ng MH, Kannan Kutty M, Selvaduray KR, Radhakrishnan AK. Reduced infiltration of regulatory T cells in tumours from mice fed daily with gamma-tocotrienol supplementation. Clin Exp Immunol 2021; 206:161-172. [PMID: 34331768 PMCID: PMC8506134 DOI: 10.1111/cei.13650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/06/2021] [Accepted: 07/11/2021] [Indexed: 01/08/2023] Open
Abstract
Gamma-tocotrienol (γT3) is an analogue of vitamin E with beneficial effects on the immune system, including immune-modulatory properties. This study reports the immune-modulatory effects of daily supplementation of γT3 on host T helper (Th) and T regulatory cell (Treg ) populations in a syngeneic mouse model of breast cancer. Female BALB/c mice were fed with either γT3 or vehicle (soy oil) for 2 weeks via oral gavage before they were inoculated with syngeneic 4T1 mouse mammary cancer cells (4T1 cells). Supplementation continued until the mice were euthanized. Mice (n = 6) were euthanized at specified time-points for various analysis (blood leucocyte, cytokine production and immunohistochemistry). Tumour volume was measured once every 7 days. Gene expression studies were carried out on tumour-specific T lymphocytes isolated from splenic cultures. Supplementation with γT3 increased CD4+ (p < 0.05), CD8+ (p < 0.05) T-cells and natural killer cells (p < 0.05) but suppressed Treg cells (p < 0.05) in peripheral blood when compared to animals fed with the vehicle. Higher interferon (IFN)-γ and lower transforming growth factor (TGF)-ꞵ levels were noted in the γT3 fed mice. Immunohistochemistry findings revealed higher infiltration of CD4+ cells, increased expression of interleukin-12 receptor-beta-2 (IL-12ꞵ2R), interleukin (IL)-24 and reduced expression of cells that express the forkhead box P3 (FoxP3) in tumours from the γT3-fed animals. Gene expression studies showed the down-regulation of seven prominent genes in splenic CD4+ T cells isolated from γT3-fed mice. Supplementation with γT3 from palm oil-induced T cell-dependent cell-mediated immune responses and suppressed T cells in the tumour microenvironment in a syngeneic mouse model of breast cancer.
Collapse
Affiliation(s)
- Shonia Subramaniam
- School of Postgraduate StudiesInternational Medical UniversityKuala LumpurMalaysia
- Product Development and Advisory ServicesMalaysian Palm Oil BoardKajangMalaysia
| | - Jeya Seela Anandha Rao
- Pathology DivisionSchool of MedicineInternational Medical UniversityKuala LumpurMalaysia
| | - Premdass Ramdas
- Division of Applied Biomedical Sciences and BiotechnologySchool of Health SciencesInternational Medical UniversityKuala LumpurMalaysia
| | - Mei Han Ng
- Engineering and ProcessingMalaysian Palm Oil BoardKajangMalaysia
| | | | | | - Ammu Kutty Radhakrishnan
- Pathology DivisionSchool of MedicineInternational Medical UniversityKuala LumpurMalaysia
- Jeffery Cheah School of Medicine and Health SciencesMonash University MalaysiaBandar SunwaySelangorMalaysia
| |
Collapse
|
203
|
Jafarzadeh A, Naseri A, Shojaie L, Nemati M, Jafarzadeh S, Bannazadeh Baghi H, Hamblin MR, Akhlagh SA, Mirzaei H. MicroRNA-155 and antiviral immune responses. Int Immunopharmacol 2021; 101:108188. [PMID: 34626873 DOI: 10.1016/j.intimp.2021.108188] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023]
Abstract
The microRNA, miR-155 regulates both adaptive and innate immune responses. In viral infections, miR-155 can affect both innate immunity (interferon response, natural killer cell activity, and macrophage polarization) and adaptive immunity (including generation of anti-viral antibodies, CD8+ cytotoxic T lymphocytes, Th17, Th2, Th1, Tfh and Treg cells). In many viral infections, the proper and timely regulation of miR-155 expression is critical for the induction of an effective anti-virus immune response and viral clearance without any harmful immunopathologic consequences. MiR-155 may also exert pro-viral effects, mainly through the inhibition of the anti-viral interferon response. Thus, dysregulated expression of miR-155 can result in virus persistence and disruption of the normal response to viral infections. This review provides a thorough discussion of the role of miR-155 in immune responses and immunopathologic reactions during viral infections, and highlights its potential as a therapeutic target.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Alma Naseri
- Department of Immunology, Islamic Azadi university of Zahedan, Zahedan, Iran
| | - Layla Shojaie
- Research Center for Liver Diseases, Keck School of Medicine, Department of Medicine, University of Southern California, Los angeles, CA, USA
| | - Maryam Nemati
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Bannazadeh Baghi
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
204
|
Luan X, Yang W, Bai X, Li H, Li H, Fan W, Zhang H, Liu W, Sun L. Cyclophilin A is a key positive and negative feedback regulator within interleukin-6 trans-signaling pathway. FASEB J 2021; 35:e21958. [PMID: 34606626 DOI: 10.1096/fj.202101044rrr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/05/2021] [Accepted: 09/13/2021] [Indexed: 11/11/2022]
Abstract
Cyclophilin A (CypA), a member of the cyclophilin family, plays a vital role in microorganismal infections, inflammatory diseases, and cancers. Interleukin-6 (IL-6) is a pleiotropic cytokine, exerting variety of effects on inflammation, immune response, hematopoiesis, and tumor proliferation. Binding of IL-6 to soluble IL-6 receptor (sIL-6R) induces pro-inflammatory trans-signaling, which has been described to be stronger than anti-inflammatory classic signaling triggered by the binding of IL-6 to membrane-bound IL-6 receptor. Here we found that upon the treatment of IL-6 and sIL-6R, CypA inhibited the ubiquitination-mediated degradation of IL-6 membrane receptor gp130 and enhanced its dimerization, thereby positively regulated the IL-6 trans-signaling and increased the expression of downstream iNOS, IL-6, and CypA. Furthermore, CypA expression could be negatively regulated by suppressor of cytokine signaling 1 (SOCS1). The SH2 and Box domains of SOCS1 interacted with CypA and promoted its K48-linked ubiquitination-mediated degradation, which inhibited the IL-6 trans-signaling pathway. Collectively, our findings reveal an important role of CypA in the positive and negative feedback regulation of the IL-6 trans-signaling pathway.
Collapse
Affiliation(s)
- Xiaohan Luan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Wenxian Yang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyuan Bai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Heqiao Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Huizi Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Wenhui Fan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - He Zhang
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Guangdong, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.,Institute of Infectious Diseases, Shenzhen Bay Laboratory, Guangdong, China.,Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China
| | - Lei Sun
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
205
|
Sobah ML, Liongue C, Ward AC. SOCS Proteins in Immunity, Inflammatory Diseases, and Immune-Related Cancer. Front Med (Lausanne) 2021; 8:727987. [PMID: 34604264 PMCID: PMC8481645 DOI: 10.3389/fmed.2021.727987] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/16/2021] [Indexed: 01/10/2023] Open
Abstract
Cytokine signaling represents one of the cornerstones of the immune system, mediating the complex responses required to facilitate appropriate immune cell development and function that supports robust immunity. It is crucial that these signals be tightly regulated, with dysregulation underpinning immune defects, including excessive inflammation, as well as contributing to various immune-related malignancies. A specialized family of proteins called suppressors of cytokine signaling (SOCS) participate in negative feedback regulation of cytokine signaling, ensuring it is appropriately restrained. The eight SOCS proteins identified regulate cytokine and other signaling pathways in unique ways. SOCS1–3 and CISH are most closely involved in the regulation of immune-related signaling, influencing processes such polarization of lymphocytes and the activation of myeloid cells by controlling signaling downstream of essential cytokines such as IL-4, IL-6, and IFN-γ. SOCS protein perturbation disrupts these processes resulting in the development of inflammatory and autoimmune conditions as well as malignancies. As a consequence, SOCS proteins are garnering increased interest as a unique avenue to treat these disorders.
Collapse
Affiliation(s)
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, VIC, Australia.,Institue of Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Geelong, VIC, Australia.,Institue of Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
206
|
Alizadeh M, Nafari A, Safarzadeh A, Veiskarami S, Almasian M, Asghar Kiani A. The Impact of EGCG and RG108 on SOCS1 Promoter DNA Methylation and Expression in U937 Leukemia Cells. Rep Biochem Mol Biol 2021; 10:455-461. [PMID: 34981023 PMCID: PMC8718778 DOI: 10.52547/rbmb.10.3.455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/15/2021] [Indexed: 04/13/2023]
Abstract
BACKGROUND The available evidence has increasingly demonstrated that a combination of genetic and epigenetic factors, such as DNA methylation, could be considered as causing leukemia. Epigenetic changes and methylation of the suppressor of the cytokine signaling 1 promoter (SOCS1) CpG region silence SOCS1 expression in cancer. In the current study, we evaluated the impact of epigallocatechin gallate (EGCG) and RG108 on SOCS1 promoter methylation and expression in U937 cells. METHODS In the current study, U937 leukemic cells were treated with EGCG and RG108 for 12, 24, 48, and 72 h and SOCS1 promoter methylation and its expression were measured by methylation-specific PCR (MSP) and quantitative real-time PCR, respectively. RESULTS The outcomes indicated that the SOCS1 promoter is methylated in U937 cells, and treatment of these cells with either EGCG or RG108 reduced its methylation. Moreover, we observed that SOCS1 expression was significantly upregulated in a time-dependent manner by both EGCG and RG108 in U937 cells compared with control cells. In the RG108-treated group at 12, 24, 48, and 72 h, SOCS1 expression was upregulated by 1, 4.2, 16.6, and 32.6 -fold respectively, and in the EGCG-treated group, by 0.5, 3.2, 10.8, and 22.3 -fold, respectively. CONCLUSION Treatment with either EGCG or RG108 reduced SOCS1 promoter methylation and increased SOCS1 expression in U937 cells in a time-dependent manner, which may play a role in leukemia therapy.
Collapse
Affiliation(s)
- Mohsen Alizadeh
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Amirhossein Nafari
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Ali Safarzadeh
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Saeed Veiskarami
- Department of animal science, Lorestan Agricultural and Natural Resources Research and Education Center, Iran
| | - Mohammad Almasian
- School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Ali Asghar Kiani
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Lorestan University of Medical Sciences, Khorramabad, Iran.
- Corresponding author: Ali Asghar Kiani; Tel: +98 9166638354; E-mail:
| |
Collapse
|
207
|
Hu J, Stojanović J, Yasamineh S, Yasamineh P, Karuppannan SK, Hussain Dowlath MJ, Serati-Nouri H. The potential use of microRNAs as a therapeutic strategy for SARS-CoV-2 infection. Arch Virol 2021; 166:2649-2672. [PMID: 34278528 PMCID: PMC8286877 DOI: 10.1007/s00705-021-05152-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/21/2021] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To date, there is no effective therapeutic approach for treating SARS-CoV-2 infections. MicroRNAs (miRNAs) have been recognized to target the viral genome directly or indirectly, thereby inhibiting viral replication. Several studies have demonstrated that host miRNAs target different sites in SARS-CoV-2 RNA and constrain the production of essential viral proteins. Furthermore, miRNAs have lower toxicity, are more immunogenic, and are more diverse than protein-based and even plasmid-DNA-based therapeutic agents. In this review, we emphasize the role of miRNAs in viral infection and their potential use as therapeutic agents against COVID-19 disease. The potential of novel miRNA delivery strategies, especially EDV™ nanocells, for targeting lung tissue for treatment of SARS-CoV-2 infection is also discussed.
Collapse
Affiliation(s)
- Jiulue Hu
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, China
| | - Jelena Stojanović
- Faculty of Mathematics and Computer Science in Belgrade, ALFA BK University, Belgrade, Serbia
| | - Saman Yasamineh
- Young Researcher and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| | - Pooneh Yasamineh
- Young Researcher and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Sathish Kumar Karuppannan
- Center for Environmental Nuclear Research, Directorate of Research and Virtual Education, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| | - Mohammed Junaid Hussain Dowlath
- Center for Environmental Nuclear Research, Directorate of Research and Virtual Education, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| | - Hamed Serati-Nouri
- Stem cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
208
|
Wang C, Liu S, Li J, Cheng Y, Wang Z, Feng T, Lu G, Wang S, Song J, Xia P, Hao L. Biological Functions of Let-7e-5p in Promoting the Differentiation of MC3T3-E1 Cells. Front Cell Dev Biol 2021; 9:671170. [PMID: 34568312 PMCID: PMC8455882 DOI: 10.3389/fcell.2021.671170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/03/2021] [Indexed: 11/18/2022] Open
Abstract
MicroRNAs let-7c and let-7f, two members of the let-7 family, were involved in regulating osteoblast differentiation and have an important role in bone formation. Let-7e-5p, which also belonged to the let-7 family, presented in the differentiation of adipose-derived stem cells and mouse embryonic stem cells. However, the role of let-7e-5p in osteoblast differentiation was unclear. Thus, this study aimed to elucidate the function of let-7e-5p in osteoblast differentiation and its mechanism. Firstly, we found that the let-7e-5p mimic promoted osteoblast differentiation but not the proliferation of MC3T3-E1 cells by positively regulating the expression levels of osteogenic-associated genes (RUNX2, OCN, OPN, and OSX), the activity of ALP, and formation of mineralized nodules. Moreover, we ascertained that the let-7e-5p mimic downregulated the post-transcriptional expression of SOCS1 by specifically binding to the 3′ untranslated region of SOCS1 mRNA. Also, let-7e-5p-induced SOCS1 downregulation increased the protein levels of p-STAT5 and IGF-1, which were both modulated by SOCS1 molecules. Furthermore, let-7e-5p abrogated the inhibition of osteogenic differentiation mediated by SOCS1 overexpression. Therefore, these results suggested that let-7e-5p regulated the differentiation of MC3T3-E1 cells through the JAK2/STAT5 pathway to upregulate IGF-1 gene expression by inhibiting SOCS1. These findings may provide a new insight into the regulatory role of let-7e-5p in osteogenic differentiation and imply the existence of a novel mechanism underlying let-7e-5p-mediated osteogenic differentiation.
Collapse
Affiliation(s)
- Chunli Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Songcai Liu
- College of Animal Science, Jilin University, Changchun, China
| | - Jiaxin Li
- College of Animal Science, Jilin University, Changchun, China
| | - Yunyun Cheng
- College of Public Health, Jilin University, Changchun, China
| | - Zhaoguo Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Tianqi Feng
- College of Animal Science, Jilin University, Changchun, China
| | - Guanhong Lu
- College of Animal Science, Jilin University, Changchun, China
| | - Siyao Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Jie Song
- College of Animal Science, Jilin University, Changchun, China
| | - Peijun Xia
- College of Animal Science, Jilin University, Changchun, China
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun, China
| |
Collapse
|
209
|
Wang R, Yang X, Chang M, Xue Z, Wang W, Bai L, Zhao S, Liu E. ORF3a Protein of Severe Acute Respiratory Syndrome Coronavirus 2 Inhibits Interferon-Activated Janus Kinase/Signal Transducer and Activator of Transcription Signaling via Elevating Suppressor of Cytokine Signaling 1. Front Microbiol 2021; 12:752597. [PMID: 34650546 PMCID: PMC8506155 DOI: 10.3389/fmicb.2021.752597] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/07/2021] [Indexed: 12/26/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has caused a crisis to global public health since its outbreak at the end of 2019. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogen of COVID-19, appears to efficiently evade the host immune responses, including interferon (IFN) signaling. Several SARS-CoV-2 viral proteins are believed to involve in the inhibition of IFN signaling. In this study, we discovered that ORF3a, an accessory protein of SARS-CoV-2, inhibited IFN-activated Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling via upregulating suppressor of cytokine signaling 1 (SOCS1), a negative regulator of cytokine signaling. ORF3a induced SOCS1 elevation in a dose- and time-dependent manner. RNAi-mediated silencing of SOCS1 efficiently abolished ORF3a-induced blockage of JAK/STAT signaling. Interestingly, we found that ORF3a also promoted the ubiquitin-proteasomal degradation of Janus kinase 2 (JAK2), an important kinase in IFN signaling. Silencing of SOCS1 by siRNA distinctly blocked ORF3a-induced JAK2 ubiquitination and degradation. These results demonstrate that ORF3a dampens IFN signaling via upregulating SOCS1, which suppressed STAT1 phosphorylation and accelerated JAK2 ubiquitin-proteasomal degradation. Furthermore, analysis of ORF3a deletion constructs showed that the middle domain of ORF3a (amino acids 70-130) was responsible for SOCS1 upregulation. These findings contribute to our understanding of the mechanism of SARS-CoV-2 antagonizing host antiviral response.
Collapse
Affiliation(s)
- Rong Wang
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | | | | | | | | | | | | | - Enqi Liu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, Xi’an, China
| |
Collapse
|
210
|
AXL Receptor in Cancer Metastasis and Drug Resistance: When Normal Functions Go Askew. Cancers (Basel) 2021; 13:cancers13194864. [PMID: 34638349 PMCID: PMC8507788 DOI: 10.3390/cancers13194864] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/15/2021] [Accepted: 09/21/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary AXL is a member of the TAM (TYRO3, AXL, MER) family of receptor tyrosine kinases. In normal physiological conditions, AXL is involved in removing dead cells and their remains, and limiting the duration of immune responses. Both functions are utilized by cancers in the course of tumour progression. Cancer cells use the AXL pathway to detect toxic environments and to activate molecular mechanisms, thereby ensuring their survival or escape from the toxic zone. AXL is instrumental in controlling genetic programs of epithelial-mesenchymal and mesenchymal-epithelial transitions, enabling cancer cells to metastasize. Additionally, AXL signaling suppresses immune responses in tumour microenvironment and thereby helps cancer cells to evade immune surveillance. The broad role of AXL in tumour biology is the reason why its inhibition sensitizes tumours to a broad spectrum of anti-cancer drugs. In this review, we outline molecular mechanisms underlying AXL function in normal tissues, and discuss how these mechanisms are adopted by cancers to become metastatic and drug-resistant. Abstract The TAM proteins TYRO3, AXL, and MER are receptor tyrosine kinases implicated in the clearance of apoptotic debris and negative regulation of innate immune responses. AXL contributes to immunosuppression by terminating the Toll-like receptor signaling in dendritic cells, and suppressing natural killer cell activity. In recent years, AXL has been intensively studied in the context of cancer. Both molecules, the receptor, and its ligand GAS6, are commonly expressed in cancer cells, as well as stromal and infiltrating immune cells. In cancer cells, the activation of AXL signaling stimulates cell survival and increases migratory and invasive potential. In cells of the tumour microenvironment, AXL pathway potentiates immune evasion. AXL has been broadly implicated in the epithelial-mesenchymal plasticity of cancer cells, a key factor in drug resistance and metastasis. Several antibody-based and small molecule AXL inhibitors have been developed and used in preclinical studies. AXL inhibition in various mouse cancer models reduced metastatic spread and improved the survival of the animals. AXL inhibitors are currently being tested in several clinical trials as monotherapy or in combination with other drugs. Here, we give a brief overview of AXL structure and regulation and discuss the normal physiological functions of TAM receptors, focusing on AXL. We present a theory of how epithelial cancers exploit AXL signaling to resist cytotoxic insults, in order to disseminate and relapse.
Collapse
|
211
|
Genetic and epigenetic insights into cutaneous T-cell lymphoma. Blood 2021; 139:15-33. [PMID: 34570882 DOI: 10.1182/blood.2019004256] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/30/2021] [Indexed: 11/20/2022] Open
Abstract
Primary cutaneous T-cell lymphomas (CTCL) constitute a heterogeneous group of non-Hodgkin T-cell lymphomas that present in the skin. In recent years significant progress has been made in the understanding of the pathogenesis of CTCL. Progress in CTCL classifications combined with technical advances, in particular next generation sequencing (NGS), enabled a more detailed analysis of the genetic and epigenetic landscape and transcriptional changes in clearly defined diagnostic entities. These studies not only demonstrated extensive heterogeneity between different CTCL subtypes but also identified recurrent alterations that are highly characteristic for diagnostic subgroups of CTCL. The identified alterations in particular involve epigenetic remodelling, cell cycle regulation, and the constitutive activation of targetable, oncogenic pathways. In this respect, aberrant JAK-STAT signaling is a recurrent theme, however not universal for all CTCL and with seemingly different underlaying causes in different entities. A number of the mutated genes identified are potentially actionable targets for the development of novel therapeutic strategies. Moreover, these studies have produced an enormous amount of information that will be critically important for the further development of improved diagnostic and prognostic biomarkers that can assist in the clinical management of CTCL patients. In the present review the main findings of these studies in relation to their functional impact on the malignant transformation process are discussed for different subtypes of CTCL.
Collapse
|
212
|
Ren A, Yin W, Miller H, Westerberg LS, Candotti F, Park CS, Lee P, Gong Q, Chen Y, Liu C. Novel Discoveries in Immune Dysregulation in Inborn Errors of Immunity. Front Immunol 2021; 12:725587. [PMID: 34512655 PMCID: PMC8429820 DOI: 10.3389/fimmu.2021.725587] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/27/2021] [Indexed: 12/19/2022] Open
Abstract
With the expansion of our knowledge on inborn errors of immunity (IEI), it gradually becomes clear that immune dysregulation plays an important part. In some cases, autoimmunity, hyperinflammation and lymphoproliferation are far more serious than infections. Thus, immune dysregulation has become significant in disease monitoring and treatment. In recent years, the wide application of whole-exome sequencing/whole-genome sequencing has tremendously promoted the discovery and further studies of new IEI. The number of discovered IEI is growing rapidly, followed by numerous studies of their pathogenesis and therapy. In this review, we focus on novel discovered primary immune dysregulation diseases, including deficiency of SLC7A7, CD122, DEF6, FERMT1, TGFB1, RIPK1, CD137, TET2 and SOCS1. We discuss their genetic mutation, symptoms and current therapeutic methods, and point out the gaps in this field.
Collapse
Affiliation(s)
- Anwen Ren
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yin
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- The Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Lisa S Westerberg
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Fabio Candotti
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Chan-Sik Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Yan Chen
- The Second Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
213
|
Zhao Y, He D, Zeng H, Luo J, Yang S, Chen J, Abdullah RK, Liu N. Expression and significance of miR-30d-5p and SOCS1 in patients with recurrent implantation failure during implantation window. Reprod Biol Endocrinol 2021; 19:138. [PMID: 34496883 PMCID: PMC8425163 DOI: 10.1186/s12958-021-00820-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/23/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Poor endometrial receptivity is a major factor that leads to recurrent implantation failure. However, the traditional method cannot accurately evaluate endometrial receptivity. Various studies have indicated that microRNAs (miRNAs) are involved in multiple processes of embryo implantation, but the role of miRNAs in endometrial receptivity in patients with recurrent implantation failure (RIF) remains elusive. In the present study, we investigated the presence of pinopodes and the roles of miR-30d-5p, suppressor of cytokine signalling 1 (SOCS1) and the leukaemia inhibitory factor (LIF) pathway in women with a history of RIF during the implantation window. METHODS Endometrial tissue samples were collected between January 2018 to June 2019 from two groups of women who underwent in vitro fertilisation and embryo transfer (IVF-ET) or frozen ET. The RIF group included 20 women who underwent ≥ 3 ETs, including a total of ≥ 4 good-quality embryos, without pregnancy, whereas the control group included 10 women who had given birth at least once in the past year. An endometrial biopsy was performed during the implantation window (LH + 7). The development of pinopodes in the endometrial biopsy samples from all groups was evaluated using scanning electron microscopy (SEM). Quantitative reverse transcription-polymerase chain reaction and western blotting were used to investigate the expression levels of miR-30d-5p, SOCS1, and the LIF pathway. RESULTS The presence of developed pinopodes decreased in patients with RIF on LH + 7. The expression level of miR-30d-5p decreased in the endometria during the implantation window of patients with RIF, whereas the mRNA and protein levels of SOCS1 were significantly higher in the RIF group than in the control group. Furthermore, a negative correlation was observed between the expression of miR-30d-5p and SOCS1 (r2 = 0.8362). In addition, a significant decrease in LIF and p-STAT3 expression was observed during the implantation window in patients with RIF. CONCLUSIONS MiR-30d-5p and SOCS1 may be potential biomarkers for endometrial receptivity. Changes in pinopode development and abnormal expression of miR-30d-5p, SOCS1 and LIF pathway in the endometrium could be the reasons for implantation failure.
Collapse
Affiliation(s)
- Yuhao Zhao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Dongmei He
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hong Zeng
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jiefeng Luo
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shuang Yang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jingjing Chen
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Raed K Abdullah
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Nenghui Liu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
214
|
Effect of (-)-Epigallocatechin Gallate on Activation of JAK/STAT Signaling Pathway by Staphylococcal Enterotoxin A. Toxins (Basel) 2021; 13:toxins13090609. [PMID: 34564613 PMCID: PMC8473440 DOI: 10.3390/toxins13090609] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 11/17/2022] Open
Abstract
Staphylococcal enterotoxin A (SEA), which is a superantigen toxin protein, binds to cytokine receptor gp130. Gp130 activates intracellular signaling pathways, including the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway. The effects of SEA on the JAK/STAT signaling pathway in mouse spleen cells were examined. After treatment with SEA, mRNA expression levels of interferon gamma (IFN-γ) and suppressor of cytokine-signaling 1 (SOCS1) increased. SEA-induced IFN-γ and SOCS1 expression were decreased by treatment with (-)-epigallocatechin gallate (EGCG). The phosphorylated STAT3, Tyr705, increased significantly in a SEA concentration-dependent manner in mouse spleen cells. Although (-)-3″-Me-EGCG did not inhibit SEA-induced phosphorylated STAT3, EGCG and (-)-4″-Me-EGCG significantly inhibited SEA-induced phosphorylated STAT3. It was thought that the hydroxyl group at position 3 of the galloyl group in the EGCG was responsible for binding to SEA and suppressing SEA-induced phosphorylation of STAT3. Through protein thermal shift assay in vitro, the binding of the gp130 receptor to SEA and the phosphorylation of STAT3 were inhibited by the interaction between EGCG and SEA. As far as we know, this is the first report to document that EGCG inhibits the binding of the gp130 receptor to SEA and the associated phosphorylation of STAT3.
Collapse
|
215
|
Körholz J, Gabrielyan A, Sowerby JM, Boschann F, Chen LS, Paul D, Brandt D, Kleymann J, Kolditz M, Toepfner N, Knöfler R, Jacobsen EM, Wolf C, Conrad K, Röber N, Lee-Kirsch MA, Smith KGC, Mundlos S, Berner R, Dalpke AH, Schuetz C, Rae W. One Gene, Many Facets: Multiple Immune Pathway Dysregulation in SOCS1 Haploinsufficiency. Front Immunol 2021; 12:680334. [PMID: 34421895 PMCID: PMC8375263 DOI: 10.3389/fimmu.2021.680334] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/13/2021] [Indexed: 01/25/2023] Open
Abstract
Background Inborn errors of immunity (IEI) present with a large phenotypic spectrum of disease, which can pose diagnostic and therapeutic challenges. Suppressor of cytokine signaling 1 (SOCS1) is a key negative regulator of cytokine signaling, and has recently been associated with a novel IEI. Of patients described to date, it is apparent that SOCS1 haploinsufficiency has a pleiotropic effect in humans. Objective We sought to investigate whether dysregulation of immune pathways, in addition to STAT1, play a role in the broad clinical manifestations of SOCS1 haploinsufficiency. Methods We assessed impacts of reduced SOCS1 expression across multiple immune cell pathways utilizing patient cells and CRISPR/Cas9 edited primary human T cells. Results SOCS1 haploinsufficiency phenotypes straddled across the International Union of Immunological Societies classifications of IEI. We found that reduced SOCS1 expression led to dysregulation of multiple intracellular pathways in immune cells. STAT1 phosphorylation is enhanced, comparably with STAT1 gain-of-function mutations, and STAT3 phosphorylation is similarly reduced with concurrent reduction of Th17 cells. Furthermore, reduced SOCS1 E3 ligase function was associated with increased FAK1 in immune cells, and increased AKT and p70 ribosomal protein S6 kinase phosphorylation. We also found Toll-like receptor responses are increased in SOCS1 haploinsufficiency patients. Conclusions SOCS1 haploinsufficiency is a pleiotropic monogenic IEI. Dysregulation of multiple immune cell pathways may explain the variable clinical phenotype associated with this new condition. Knowledge of these additional dysregulated immune pathways is important when considering the optimum management for SOCS1 haploinsufficient patients.
Collapse
Affiliation(s)
- Julia Körholz
- Department of Pediatrics, University Hospital and Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany.,UniversitätsCentrum für seltene Erkrankungen, Medizinische Fakultät Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Anastasia Gabrielyan
- Department of Pediatrics, University Hospital and Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - John M Sowerby
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom.,Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Felix Boschann
- Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Lan-Sun Chen
- Institute of Medical Microbiology and Virology, Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Diana Paul
- Department of Pediatrics, University Hospital and Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - David Brandt
- Department of Pediatrics, University Hospital and Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Janina Kleymann
- Department of Internal Medicine, Pneumology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Martin Kolditz
- Department of Internal Medicine, Pneumology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nicole Toepfner
- Department of Pediatrics, University Hospital and Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Ralf Knöfler
- Department of Pediatrics, University Hospital and Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Christine Wolf
- Department of Pediatrics, University Hospital and Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Karsten Conrad
- Institute of Immunology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nadja Röber
- Institute of Immunology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Min Ae Lee-Kirsch
- Department of Pediatrics, University Hospital and Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany.,UniversitätsCentrum für seltene Erkrankungen, Medizinische Fakultät Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom.,Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Stefan Mundlos
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Max Planck Institute for Molecular Genetics, Research Group (RG) Development and Disease, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Reinhard Berner
- Department of Pediatrics, University Hospital and Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany.,UniversitätsCentrum für seltene Erkrankungen, Medizinische Fakultät Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Alexander H Dalpke
- Institute of Medical Microbiology and Virology, Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Catharina Schuetz
- Department of Pediatrics, University Hospital and Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany.,UniversitätsCentrum für seltene Erkrankungen, Medizinische Fakultät Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - William Rae
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom.,Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
216
|
Yoshimura A, Ito M, Mise-Omata S, Ando M. SOCS: negative regulators of cytokine signaling for immune tolerance. Int Immunol 2021; 33:711-716. [PMID: 34415326 DOI: 10.1093/intimm/dxab055] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/18/2021] [Indexed: 11/14/2022] Open
Abstract
Cytokines are important intercellular communication tools for immunity. Many cytokines promote gene transcription and proliferation through the JAK/STAT (Janus kinase / signal transducers and activators of transcription) and the Ras/ERK (GDP/GTP-binding rat sarcoma protein / extracellular signal-regulated kinase) pathways, and these signaling pathways are tightly regulated. The SOCS (suppressor of cytokine signaling) family are representative negative regulators of JAK/STAT-mediated cytokine signaling and regulate the differentiation and function of T cells, thus being involved in immune tolerance. Human genetic analysis has shown that SOCS family members are strongly associated with autoimmune diseases, allergy and tumorigenesis. SOCS family proteins also function as immune-checkpoint molecules that contribute to the unresponsiveness of T cells to cytokines.
Collapse
Affiliation(s)
- Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Shinjyuku-ku, Tokyo, Japan
| | - Minako Ito
- Medical Institute of Bioregulation Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Setsuko Mise-Omata
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Shinjyuku-ku, Tokyo, Japan
| | - Makoto Ando
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Shinjyuku-ku, Tokyo, Japan
| |
Collapse
|
217
|
Moser B, Edtmayer S, Witalisz-Siepracka A, Stoiber D. The Ups and Downs of STAT Inhibition in Acute Myeloid Leukemia. Biomedicines 2021; 9:1051. [PMID: 34440253 PMCID: PMC8392322 DOI: 10.3390/biomedicines9081051] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 01/03/2023] Open
Abstract
Aberrant Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling is implicated in the pathogenesis of acute myeloid leukemia (AML), a highly heterogeneous hematopoietic malignancy. The management of AML is complex and despite impressive efforts into better understanding its underlying molecular mechanisms, survival rates in the elderly have not shown a substantial improvement over the past decades. This is particularly due to the heterogeneity of AML and the need for personalized approaches. Due to the crucial role of the deregulated JAK-STAT signaling in AML, selective targeting of the JAK-STAT pathway, particularly constitutively activated STAT3 and STAT5 and their associated upstream JAKs, is of great interest. This strategy has shown promising results in vitro and in vivo with several compounds having reached clinical trials. Here, we summarize recent FDA approvals and current potential clinically relevant inhibitors for AML patients targeting JAK and STAT proteins. This review underlines the need for detailed cytogenetic analysis and additional assessment of JAK-STAT pathway activation. It highlights the ongoing development of new JAK-STAT inhibitors with better disease specificity, which opens up new avenues for improved disease management.
Collapse
Affiliation(s)
| | | | | | - Dagmar Stoiber
- Department of Pharmacology, Physiology and Microbiology, Division Pharmacology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (B.M.); (S.E.); (A.W.-S.)
| |
Collapse
|
218
|
The Role of the IL-6 Cytokine Family in Epithelial-Mesenchymal Plasticity in Cancer Progression. Int J Mol Sci 2021; 22:ijms22158334. [PMID: 34361105 PMCID: PMC8347315 DOI: 10.3390/ijms22158334] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/12/2021] [Accepted: 07/28/2021] [Indexed: 02/07/2023] Open
Abstract
Epithelial–mesenchymal plasticity (EMP) plays critical roles during embryonic development, wound repair, fibrosis, inflammation and cancer. During cancer progression, EMP results in heterogeneous and dynamic populations of cells with mixed epithelial and mesenchymal characteristics, which are required for local invasion and metastatic dissemination. Cancer development is associated with an inflammatory microenvironment characterized by the accumulation of multiple immune cells and pro-inflammatory mediators, such as cytokines and chemokines. Cytokines from the interleukin 6 (IL-6) family play fundamental roles in mediating tumour-promoting inflammation within the tumour microenvironment, and have been associated with chronic inflammation, autoimmunity, infectious diseases and cancer, where some members often act as diagnostic or prognostic biomarkers. All IL-6 family members signal through the Janus kinase (JAK)–signal transducer and activator of transcription (STAT) pathway and are able to activate a wide array of signalling pathways and transcription factors. In general, IL-6 cytokines activate EMP processes, fostering the acquisition of mesenchymal features in cancer cells. However, this effect may be highly context dependent. This review will summarise all the relevant literature related to all members of the IL-6 family and EMP, although it is mainly focused on IL-6 and oncostatin M (OSM), the family members that have been more extensively studied.
Collapse
|
219
|
Nohara S, Yamamoto M, Yasukawa H, Nagata T, Takahashi J, Shimozono K, Yanai T, Sasaki T, Okabe K, Shibata T, Akagaki D, Mawatari K, Fukumoto Y. SOCS3 deficiency in cardiomyocytes elevates sensitivity of ischemic preconditioning that synergistically ameliorates myocardial ischemia reperfusion injury. PLoS One 2021; 16:e0254712. [PMID: 34292971 PMCID: PMC8297769 DOI: 10.1371/journal.pone.0254712] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/01/2021] [Indexed: 11/19/2022] Open
Abstract
Ischemic preconditioning (IPC) is the most powerful endogenous cardioprotective form of cellular adaptation. However, the inhibitory or augmenting mechanism underlying cardioprotection via IPC remains largely unknown. Suppressor of cytokine signaling-3 (SOCS3) is a cytokine-inducible potent negative feedback regulator of the signal transducer and activator of transcription-3 (STAT3) signaling pathway. Here, we aimed to determine whether cardiac SOCS3 deficiency and IPC would synergistically reduce infarct size after myocardial ischemia reperfusion injury. We evaluated STAT3 activation and SOCS3 induction after ischemic conditioning (IC) using western blot analysis and real-time PCR, and found that myocardial IC alone transiently activated myocardial STAT3 and correspondingly induced SOCS3 expression in wild-type mice. Compared with wild-type mice, cardiac-specific SOCS3 knockout (SOCS3-CKO) mice showed significantly greater and more sustained IC-induced STAT3 activation. Following ischemia reperfusion, IPC substantially reduced myocardial infarct size and significantly enhanced STAT3 phosphorylation in SOCS3-CKO mice compared to in wild-type mice. Real-time PCR array analysis revealed that SOCS3-CKO mice after IC exhibited significantly increased expressions of several anti-apoptotic genes and SAFE pathway-related genes. Moreover, real-time PCR analysis revealed that myocardial IC alone rapidly induced expression of the STAT3-activating cytokine erythropoietin in the kidney at 1 h post-IC. We also found that the circulating erythropoietin level was promptly increased at 1 h after myocardial IC. Myocardial SOCS3 deficiency and IPC exert synergistic effects in the prevention of myocardial injury after ischemia reperfusion. Our present results suggest that myocardial SOCS3 is a potent inhibitor of IPC-induced cardioprotection, and that myocardial SOCS3 inhibition augment IPC-mediated cardioprotection during ischemia reperfusion injury.
Collapse
Affiliation(s)
- Shoichiro Nohara
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Mai Yamamoto
- Cardiovascular Research Institute, Kurume University, Kurume, Japan
| | - Hideo Yasukawa
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
- * E-mail:
| | - Takanobu Nagata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Jinya Takahashi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Koutatsu Shimozono
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Toshiyuki Yanai
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tomoko Sasaki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Kota Okabe
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tatsuhiro Shibata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Daiki Akagaki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Kazutoshi Mawatari
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Yoshihiro Fukumoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
- Cardiovascular Research Institute, Kurume University, Kurume, Japan
| |
Collapse
|
220
|
Chapman JR, Bouska AC, Zhang W, Alderuccio JP, Lossos IS, Rimsza LM, Maguire A, Yi S, Chan WC, Vega F, Song JY. EBV-positive HIV-associated diffuse large B cell lymphomas are characterized by JAK/STAT (STAT3) pathway mutations and unique clinicopathologic features. Br J Haematol 2021; 194:870-878. [PMID: 34272731 DOI: 10.1111/bjh.17708] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/22/2021] [Accepted: 06/29/2021] [Indexed: 12/20/2022]
Abstract
Even in the era of highly active combination antiretroviral therapy (cART), patients with HIV have a disproportionate risk of developing aggressive lymphomas that are frequently Epstein-Barr virus (EBV)-related. Here, we investigate HIV-associated diffuse large B-cell lymphoma (HIV-DLBCL) and compare EBV-positive and EBV-negative cases. HIV-DLBCL were identified from two academic medical centres and characterised by immunohistochemistry, EBV status, fluorescence in situ hybridisation, cell of origin determination by gene expression profiling, and targeted deep sequencing using a custom mutation panel of 334 genes. We also applied the Lymphgen tool to determine the genetic subtype of each case. Thirty HIV-DLBCL were identified, with a median patient age of 46 years and male predominance (5:1). Thirteen cases (48%) were EBV-positive and 14 (52%) EBV-negative. Nine of the 16 tested cases (56%) had MYC rearrangement, three (19%) had BCL6 (two of which were double hit MYC/BCL6) and none had BCL2 rearrangements. Using the Lymphgen tool, half of the cases (15) were classified as other. All HIV-DLBCL showed mutational abnormalities, the most frequent being TP53 (37%), MYC (30%), STAT3 (27%), HIST1H1E (23%), EP300 (20%), TET2 (20%), SOCS1 (17%) and SGK1 (17%). EBV-negative cases were mostly of germinal centre B-cell (GCB) origin (62%), showed more frequent mutations per case (a median of 13·5/case) and significant enrichment of TP53 (57% vs. 15%; P = 0·046), SGK1 (36% vs. 0%; P = 0·04), EP300 (43% vs. 0%; P = 0·02) and histone-modifying gene (e.g. HIST1H1E, HIST1H1D, 79% vs. 31%; P = 0·02) mutations. EBV-positive cases were mostly of non-GCB origin (70%), with fewer mutations per case (median 8/case; P = 0·007), and these tumours were enriched for STAT3 mutations (P = 0·10). EBV-positive cases had a higher frequency of MYC mutations but the difference was not significant (36% vs. 15%; P = 0·38). EBV-association was more frequent in HIV-DLBCLs, arising in patients with lower CD4 counts at diagnosis (median 46·5 vs. 101, P = 0·018). In the era of cART, approximately half of HIV-DLBCL are EBV-related. HIV-DLBCL are enriched for MYC rearrangements, MYC mutations and generally lack BCL2 rearrangements, regardless of EBV status. Among HIV-DLBCL, tumours that are EBV-negative and EBV-positive appear to have important differences, the latter arising in context of lower CD4 count, showing frequent non-GCB origin, lower mutation burden and recurrent STAT3 mutations.
Collapse
Affiliation(s)
- Jennifer R Chapman
- Division of Hematopathology, Department of Pathology, University of Miami and Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Alyssa C Bouska
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Weiwei Zhang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Juan Pablo Alderuccio
- Division of Hematology, Department of Medicine, University of Miami and Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Izidore S Lossos
- Division of Hematology, Department of Medicine, University of Miami and Sylvester Comprehensive Cancer Center, Miami, FL, USA.,Department of Molecular and Cellular Pharmacology, University of Miami, Miami, FL, USA
| | - Lisa M Rimsza
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Alanna Maguire
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Shuhua Yi
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Wing C Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Francisco Vega
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Joo Y Song
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
221
|
Prakash A, Saxena VK, Kumar R, Tomar S, Singh MK, Singh G. Differential gene expression in liver of colored broiler chicken divergently selected for residual feed intake. Trop Anim Health Prod 2021; 53:403. [PMID: 34268607 DOI: 10.1007/s11250-021-02844-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 07/02/2021] [Indexed: 10/20/2022]
Abstract
Feed constitutes about 60-70% of the total cost of poultry production. So maximizing the feed efficiency will reduce production cost. The rapid growth in the juvenile period is essential to achieve higher body weight. Therefore, identifying the genes and pathways involved in rapid growth at an early age with a lesser requirement of feed is of utmost importance to further economize the broiler production. The efficiency of feed utilization was measured using RFI (residual feed intake). The present study aimed to estimate the RFI (0-5 week) in a population of indigenously developed colored broiler sire line chicken as well as identifying the differentially expressed genes influencing RFI in high and low RFI groups. The liver samples of high and low RFI broiler chicken aged 35 days were used for microarray analysis. A total of 2798 differentially expressed genes (DEGs) were identified, out of which 913 genes were downregulated and 1885 were upregulated. The fold change varied from - 475.17 to 552.94. A subset of genes was confirmed by qRT-PCR, and outcomes were matched well with microarray data. In the functional annotation study of DEGs, the highest significant GO (Gene Ontology) terms in the biological process included protein transport, protein localization, regulation of apoptosis, and mitochondrial transport. Gene network analysis of these DEGs plays an important role to understand the interaction among genes. Study of the important genes which were differentially expressed and the related molecular pathways in this population may hold the potential for future breeding strategies for augmenting feed efficiency.
Collapse
Affiliation(s)
- A Prakash
- College of Veterinary Science, GADVASU, Rampura Phul, Bathinda, Punjab, India.
| | - V K Saxena
- Division of Avian Genetics and Breeding, Central Avian Research Institute - Indian Council of Agricultural Research, Izatnagar, Bareilly, 243122, Uttar Pradesh, India
| | - Ravi Kumar
- Department of Animal Biotechnology, National Institute of Animal Biotechnology, Hyderabad, 500075, Telangana, India
| | - S Tomar
- Division of Avian Genetics and Breeding, Central Avian Research Institute - Indian Council of Agricultural Research, Izatnagar, Bareilly, 243122, Uttar Pradesh, India
| | - M K Singh
- COVS, DUVASU, Mathura, Uttar Pradesh, India
| | - Gagandeep Singh
- College of Veterinary Science, GADVASU, Rampura Phul, Bathinda, Punjab, India
| |
Collapse
|
222
|
Froggatt HM, Harding AT, Chaparian RR, Heaton NS. ETV7 limits antiviral gene expression and control of influenza viruses. Sci Signal 2021; 14:14/691/eabe1194. [PMID: 34257104 DOI: 10.1126/scisignal.abe1194] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The type I interferon (IFN) response is an important component of the innate immune response to viral infection. Precise control of IFN responses is critical because insufficient expression of IFN-stimulated genes (ISGs) can lead to a failure to restrict viral spread, whereas excessive ISG activation can result in IFN-related pathologies. Although both positive and negative regulatory factors control the magnitude and duration of IFN signaling, it is also appreciated that several ISGs regulate aspects of the IFN response themselves. In this study, we performed a CRISPR activation screen to identify previously unknown regulators of the type I IFN response. We identified the strongly induced ISG encoding ETS variant transcription factor 7 (ETV7) as a negative regulator of the type I IFN response. However, ETV7 did not uniformly suppress ISG transcription. Instead, ETV7 preferentially targeted a subset of antiviral ISGs that were particularly important for IFN-mediated control of influenza viruses. Together, our data assign a function for ETV7 as an IFN response regulator and also identify ETV7 as a potential therapeutic target to increase innate antiviral responses and enhance IFN-based antiviral therapies.
Collapse
Affiliation(s)
- Heather M Froggatt
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Alfred T Harding
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ryan R Chaparian
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Nicholas S Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
223
|
Hromadová D, Elewaut D, Inman RD, Strobl B, Gracey E. From Science to Success? Targeting Tyrosine Kinase 2 in Spondyloarthritis and Related Chronic Inflammatory Diseases. Front Genet 2021; 12:685280. [PMID: 34290741 PMCID: PMC8287328 DOI: 10.3389/fgene.2021.685280] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/02/2021] [Indexed: 12/16/2022] Open
Abstract
Spondyloarthritis (SpA) is a family of inflammatory arthritic diseases, which includes the prototypes of psoriatic arthritis and ankylosing spondylitis. SpA is commonly associated with systemic inflammatory diseases, such as psoriasis and inflammatory bowel disease. Immunological studies, murine models and the genetics of SpA all indicate a pathogenic role for the IL-23/IL-17 axis. Therapeutics targeting the IL-23/IL-17 pathway are successful at providing symptomatic relief, but may not provide complete protection against progression of arthritis. Thus there is still tremendous interest in the discovery of novel therapeutic targets for SpA. Tyrosine kinase 2 (TYK2) is a member of the Janus kinases, which mediate intracellular signaling of cytokines via signal transducer and activator of transcription (STAT) activation. TYK2 plays a crucial role in mediating IL-23 receptor signaling and STAT3 activation. A plethora of natural mutations in and around TYK2 have provided a wealth of data to associate this kinase with autoimmune/autoinflammatory diseases in humans. Induced and natural mutations in murine Tyk2 largely support human data; however, key inter-species differences exist, which means extrapolation of data from murine models to humans needs to be done with caution. Despite these reservations, novel selective TYK2 inhibitors are now proving successful in advanced clinical trials of inflammatory diseases. In this review, we will discuss TYK2 from basic biology to therapeutic targeting, with an emphasis on studies in SpA. Seminal studies uncovering the basic science of TYK2 have provided sound foundations for targeting it in SpA and related inflammatory diseases. TYK2 inhibitors may well be the next blockbuster therapeutic for SpA.
Collapse
Affiliation(s)
- Dominika Hromadová
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dirk Elewaut
- Molecular Immunology and Inflammation Unit, VIB Centre for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Robert D. Inman
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Departments of Medicine and Immunology, University of Toronto, Toronto, ON, Canada
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eric Gracey
- Molecular Immunology and Inflammation Unit, VIB Centre for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
224
|
Combination of CRISPR/Cas9 System and CAR-T Cell Therapy: A New Era for Refractory and Relapsed Hematological Malignancies. Curr Med Sci 2021; 41:420-430. [PMID: 34218353 DOI: 10.1007/s11596-021-2391-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy is the novel treatment strategy for hematological malignancies such as acute lymphoblastic leukemia (ALL), lymphoma and multiple myeloma. However, treatment-related toxicities such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) have become significant hurdles to CAR-T treatment. Multiple strategies were established to alter the CAR structure on the genomic level to improve efficacy and reduce toxicities. Recently, the innovative gene-editing technology-clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated nuclease9 (Cas9) system, which particularly exhibits preponderance in knock-in and knockout at specific sites, is widely utilized to manufacture CAR-T products. The application of CRISPR/Cas9 to CAR-T cell therapy has shown promising clinical results with minimal toxicity. In this review, we summarized the past achievements of CRISPR/Cas9 in CAR-T therapy and focused on the potential CAR-T targets.
Collapse
|
225
|
MaruYama T, Kobayashi S, Nakatsukasa H, Moritoki Y, Taguchi D, Sunagawa Y, Morimoto T, Asao A, Jin W, Owada Y, Ishii N, Iwabuchi Y, Yoshimura A, Chen W, Shibata H. The Curcumin Analog GO-Y030 Controls the Generation and Stability of Regulatory T Cells. Front Immunol 2021; 12:687669. [PMID: 34248973 PMCID: PMC8261301 DOI: 10.3389/fimmu.2021.687669] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/31/2021] [Indexed: 12/20/2022] Open
Abstract
Regulatory T cells (Tregs) play a crucial role in preventing antitumor immune responses in cancer tissues. Cancer tissues produce large amounts of transforming growth factor beta (TGF-β), which promotes the generation of Foxp3+ Tregs from naïve CD4+ T cells in the local tumor microenvironment. TGF-β activates nuclear factor kappa B (NF-κB)/p300 and SMAD signaling, which increases the number of acetylated histones at the Foxp3 locus and induces Foxp3 gene expression. TGF-β also helps stabilize Foxp3 expression. The curcumin analog and antitumor agent, GO-Y030, prevented the TGF-β-induced generation of Tregs by preventing p300 from accelerating NF-κB-induced Foxp3 expression. Moreover, the addition of GO-Y030 resulted in a significant reduction in the number of acetylated histones at the Foxp3 promoter and at the conserved noncoding sequence 1 regions that are generated in response to TGF-β. In vivo tumor models demonstrated that GO-Y030-treatment prevented tumor growth and reduced the Foxp3+ Tregs population in tumor-infiltrating lymphocytes. Therefore, GO-Y030 exerts a potent anticancer effect by controlling Treg generation and stability.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Phytogenic/pharmacology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Coculture Techniques
- Curcumin/analogs & derivatives
- Curcumin/pharmacology
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Lymphocyte Activation/drug effects
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Mice, Inbred C57BL
- Mice, Transgenic
- NF-kappa B/metabolism
- Skin Neoplasms/drug therapy
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Burden/drug effects
- p300-CBP Transcription Factors/metabolism
- Mice
Collapse
Affiliation(s)
- Takashi MaruYama
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institute of Health, Bethesda, MS, United States
- Department of Immunology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Shuhei Kobayashi
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hiroko Nakatsukasa
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Moritoki
- Department of General Internal Medicine and Clinical Laboratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Daiki Taguchi
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Yoichi Sunagawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Tatsuya Morimoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Atsuko Asao
- Department of Microbiology and Immunology, Graduate School of Medicine, Tohoku University, Miyagi, Japan
| | - Wenwen Jin
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institute of Health, Bethesda, MS, United States
| | - Yuji Owada
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Graduate School of Medicine, Tohoku University, Miyagi, Japan
| | - Yoshiharu Iwabuchi
- Department of Organic Chemistry, Graduate School of Pharmaceutics, Tohoku University, Miyagi, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institute of Health, Bethesda, MS, United States
| | - Hiroyuki Shibata
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Akita, Japan
| |
Collapse
|
226
|
Gocher AM, Workman CJ, Vignali DAA. Interferon-γ: teammate or opponent in the tumour microenvironment? Nat Rev Immunol 2021; 22:158-172. [PMID: 34155388 DOI: 10.1038/s41577-021-00566-3] [Citation(s) in RCA: 328] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy offers substantive benefit to patients with various tumour types, in some cases leading to complete tumour clearance. However, many patients do not respond to immunotherapy, galvanizing the field to define the mechanisms of pre-existing and acquired resistance. Interferon-γ (IFNγ) is a cytokine that has both protumour and antitumour activities, suggesting that it may serve as a nexus for responsiveness to immunotherapy. Many cancer immunotherapies and chemotherapies induce IFNγ production by various cell types, including activated T cells and natural killer cells. Patients resistant to these therapies commonly have molecular aberrations in the IFNγ signalling pathway or express resistance molecules driven by IFNγ. Given that all nucleated cells can respond to IFNγ, the functional consequences of IFNγ production need to be carefully dissected on a cell-by-cell basis. Here, we review the cells that produce IFNγ and the different effects of IFNγ in the tumour microenvironment, highlighting the pleiotropic nature of this multifunctional and abundant cytokine.
Collapse
Affiliation(s)
- Angela M Gocher
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA. .,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
227
|
Sanguedolce F, Zanelli M, Zizzo M, Luminari S, Martino G, Soriano A, Ricci L, Caprera C, Ascani S. Indolent T-Cell Lymphoproliferative Disorders of the Gastrointestinal Tract (iTLPD-GI): A Review. Cancers (Basel) 2021; 13:cancers13112790. [PMID: 34205136 PMCID: PMC8199971 DOI: 10.3390/cancers13112790] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary This review aims to better define the clinical, pathological, and molecular features of the novel lymphoproliferative disease termed “indolent T-cell lymphoproliferative disorder of the gastro-intestinal tract (iTLPD-GI)”, to discuss potential pitfalls in differentiating this entity from other neoplastic and non-neoplastic disorders arising at the same site, and to point out a biomarker-based approach to the diagnosis. Abstract iTLPD-GI is a low-grade clonal T-cell lymphoproliferative disease arising in GI organs. It is an uncommon disease, and only recently has it been enlisted as a distinct provisional entity in the current WHO Classification. Data from the literature disclose high heterogeneity in terms of pathological and molecular features; on the other hand, establishing an accurate diagnosis of iTLPD-GI is of pivotal importance, since treatment options are different from that of other, more frequent lymphomas that arise in the gastrointestinal tract. In this review, we aimed to better define this novel entity, and to identify useful diagnostic biomarkers; moreover, we provide a biomarker-based approach to the diagnosis and describe the most common issues in differentiating iTLPD-GI from other neoplastic and non-neoplastic disorders.
Collapse
Affiliation(s)
- Francesca Sanguedolce
- Pathology Unit, Policlinico Riuniti, University of Foggia, 71121 Foggia, Italy
- Correspondence: ; Tel.: +39-881-736-315
| | - Magda Zanelli
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy;
| | - Maurizio Zizzo
- Surgical Oncology Unit, Azienda USL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy;
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Stefano Luminari
- Hematology Unit, Azienda USL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy;
| | - Giovanni Martino
- Hematology Unit, University of Perugia, CREO Perugia, 06124 Perugia, Italy;
| | - Alessandra Soriano
- Gastroenterology Unit, Azienda USL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy;
| | - Linda Ricci
- Pathology Unit, Azienda Ospedaliera S. Maria di Terni, University of Perugia, 05100 Terni, Italy; (L.R.); (C.C.); (S.A.)
| | - Cecilia Caprera
- Pathology Unit, Azienda Ospedaliera S. Maria di Terni, University of Perugia, 05100 Terni, Italy; (L.R.); (C.C.); (S.A.)
| | - Stefano Ascani
- Pathology Unit, Azienda Ospedaliera S. Maria di Terni, University of Perugia, 05100 Terni, Italy; (L.R.); (C.C.); (S.A.)
| |
Collapse
|
228
|
Satarker S, Tom AA, Shaji RA, Alosious A, Luvis M, Nampoothiri M. JAK-STAT Pathway Inhibition and their Implications in COVID-19 Therapy. Postgrad Med 2021; 133:489-507. [PMID: 33245005 PMCID: PMC7784782 DOI: 10.1080/00325481.2020.1855921] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
As the incidence of COVID-19 increases with time, more and more efforts are made to pave a way out for the therapeutic strategies to deal with the disease progression. Inflammation being a significant influencer in COVID-19 patients, it drives our focus onto the signaling cascades of the JAK/STAT pathway. JAK phosphorylation mediated by cytokine receptor activation leads to phosphorylation of STATs that translocate into the nucleus to translate for inflammatory mediators. The SARS-CoV-2 structural proteins like spike, nucleocapsid, membrane and envelope proteins along with the non- structural proteins 1-16 including proteases like 3CL pro and PLpro promote its entry and survival in hosts. The SARS-CoV-2 infection triggers inflammation via the JAK/STAT pathway leading to recruitment of pneumocytes, endothelial cells, macrophages, monocytes, lymphocytes, natural killer cells and dendritic cells progressing towards cytokine storm. This produces various inflammatory markers in the host that determine the disease severity. The JAK/STAT signaling also mediates immune responses via B cell and T cell differentiation.With an attempt to reduce excessive inflammation, JAK/STAT inhibitors like Ruxolitinib, Baricitinib, Tofacitinib have been employed that mediate its actions via suppressors of cytokine signaling, cytokine inducible SH2 containing protein, Protein inhibitor of activated STAT and protein tyrosine phosphatases. Even though they are implicated with multiple adverse effects, the regulatory authorities have supported its use, and numerous clinical trials are in progress to prove their safety and efficacy. On the contrary, the exact mechanism of JAK/STAT inhibition at molecular levels remains speculative for which further investigations are required.
Collapse
Affiliation(s)
- Sairaj Satarker
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Antriya Annie Tom
- Department of Pharmacy Practice, Nirmala College of Pharmacy, Muvattupuzha, Kerala, India
| | - Roshitha Ann Shaji
- Department of Pharmacy Practice, Nirmala College of Pharmacy, Muvattupuzha, Kerala, India
| | - Aaja Alosious
- Department of Pharmacy Practice, Nirmala College of Pharmacy, Muvattupuzha, Kerala, India
| | - Mariya Luvis
- Department of Pharmacy Practice, Nirmala College of Pharmacy, Muvattupuzha, Kerala, India
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
229
|
Morichika K, Karube K, Sakihama S, Watanabe R, Kawaki M, Nishi Y, Nakachi S, Okamoto S, Takahara T, Satou A, Shimada S, Shimada K, Tsuzuki T, Fukushima T, Morishima S, Masuzaki H. The Positivity of Phosphorylated STAT3 Is a Novel Marker for Favorable Prognosis in Germinal Center B-Cell Type of Diffuse Large B-Cell Lymphoma. Am J Surg Pathol 2021; 45:832-840. [PMID: 33899787 DOI: 10.1097/pas.0000000000001691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
On the basis of immunohistochemistry, diffuse large B-cell lymphoma (DLBCL) is categorized as a germinal center B-cell (GCB) or non-GCB subtype. Recent integrated genomic analyses have highlighted the importance of the JAK-STAT3 pathway in the molecular pathogenesis of DLBCL. However, its relevance to clinical outcomes remains controversial. Therefore, we evaluated the extent of the nuclear expression of phosphorylated STAT3 (pSTAT3), a surrogate marker of signal transducer and activator of transcription 3 (STAT3) activation, by immunohistochemistry. We also analyzed the potential relationship between pSTAT3 positivity (defined as ≥40% positive neoplastic cells) and clinicopathologic characteristics in 294 patients with DLBCL. pSTAT3 was detected in 122 patients (42%), with a higher rate in the non-GCB subtype than in the GCB subtype (57% vs. 28%, P<0.001). Factors potentially activating STAT3, MYD88L265P, and Epstein-Barr virus-encoded small RNA were identified in the pSTAT3-positive non-GCB subtype, whereas the pSTAT3-positive GCB subtype often showed STAT3 mutations and lacked EZH2 mutations and the rearrangements of BCL2 and MYC. Multivariate analyses revealed that the pSTAT3-positive GCB subtype showed a favorable prognosis (HR: 0.17; 95% confidence interval, 0.04-0.7; P=0.014). These findings suggest that pSTAT3 positivity may have a unique impact on the clinicopathologic characteristics of DLBCL, making it a promising novel marker for the favorable prognosis of patients with the GCB subtype.
Collapse
MESH Headings
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- DNA Mutational Analysis
- Enhancer of Zeste Homolog 2 Protein/genetics
- Female
- Gene Rearrangement
- Herpesvirus 4, Human/genetics
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Japan
- Lymphoma, Large B-Cell, Diffuse/chemistry
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/virology
- Male
- Middle Aged
- Mutation
- Myeloid Differentiation Factor 88/genetics
- Phosphorylation
- Prognosis
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-myc/genetics
- RNA, Viral/genetics
- STAT3 Transcription Factor/analysis
- STAT3 Transcription Factor/genetics
- Suppressor of Cytokine Signaling 1 Protein/genetics
Collapse
Affiliation(s)
- Kazuho Morichika
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine)
| | - Kennosuke Karube
- Department of Pathology and Cell Biology, Graduate School of Medicine
| | - Shugo Sakihama
- Department of Pathology and Cell Biology, Graduate School of Medicine
| | | | | | - Yukiko Nishi
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine)
| | - Sawako Nakachi
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine)
| | - Shiki Okamoto
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine)
| | - Taishi Takahara
- Department of Surgical Pathology, Aichi Medical University Hospital
| | - Akira Satou
- Department of Surgical Pathology, Aichi Medical University Hospital
| | | | - Kazuyuki Shimada
- Hematology and Oncology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University Hospital
| | - Takuya Fukushima
- Laboratory of Hematoimmunology, School of Health Sciences, Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Satoko Morishima
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine)
| | - Hiroaki Masuzaki
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine)
| |
Collapse
|
230
|
Having an Old Friend for Dinner: The Interplay between Apoptotic Cells and Efferocytes. Cells 2021; 10:cells10051265. [PMID: 34065321 PMCID: PMC8161178 DOI: 10.3390/cells10051265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/02/2023] Open
Abstract
Apoptosis, the programmed and intentional death of senescent, damaged, or otherwise superfluous cells, is the natural end-point for most cells within multicellular organisms. Apoptotic cells are not inherently damaging, but if left unattended, they can lyse through secondary necrosis. The resulting release of intracellular contents drives inflammation in the surrounding tissue and can lead to autoimmunity. These negative consequences of secondary necrosis are avoided by efferocytosis—the phagocytic clearance of apoptotic cells. Efferocytosis is a product of both apoptotic cells and efferocyte mechanisms, which cooperate to ensure the rapid and complete removal of apoptotic cells. Herein, we review the processes used by apoptotic cells to ensure their timely removal, and the receptors, signaling, and cellular processes used by efferocytes for efferocytosis, with a focus on the receptors and signaling driving this process.
Collapse
|
231
|
Glassman CR, Su L, Majri-Morrison SS, Winkelmann H, Mo F, Li P, Pérez-Cruz M, Ho PP, Koliesnik I, Nagy N, Hnizdilova T, Picton LK, Kovar M, Bollyky P, Steinman L, Meyer E, Piehler J, Leonard WJ, Garcia KC. Calibration of cell-intrinsic interleukin-2 response thresholds guides design of a regulatory T cell biased agonist. eLife 2021; 10:e65777. [PMID: 34003116 PMCID: PMC8131104 DOI: 10.7554/elife.65777] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/30/2021] [Indexed: 12/15/2022] Open
Abstract
Interleukin-2 is a pleiotropic cytokine that mediates both pro- and anti-inflammatory functions. Immune cells naturally differ in their sensitivity to IL-2 due to cell type and activation state-dependent expression of receptors and signaling pathway components. To probe differences in IL-2 signaling across cell types, we used structure-based design to create and profile a series of IL-2 variants with the capacity to titrate maximum signal strength in fine increments. One of these partial agonists, IL-2-REH, specifically expanded Foxp3+ regulatory T cells with reduced activity on CD8+ T cells due to cell type-intrinsic differences in IL-2 signaling. IL-2-REH elicited cell type-dependent differences in gene expression and provided mixed therapeutic results: showing benefit in the in vivo mouse dextran sulfate sodium (DSS) model of colitis, but no therapeutic efficacy in a transfer colitis model. Our findings show that cytokine partial agonists can be used to calibrate intrinsic differences in response thresholds across responding cell types to narrow pleiotropic actions, which may be generalizable to other cytokine and growth factor systems.
Collapse
Affiliation(s)
- Caleb R Glassman
- Department of Molecular and Cellular Physiology, Stanford University School of MedicineStanfordUnited States
- Immunology Graduate Program, Stanford University School of MedicineStanfordUnited States
- Department of Structural Biology, Stanford University School of MedicineStanfordUnited States
| | - Leon Su
- Department of Molecular and Cellular Physiology, Stanford University School of MedicineStanfordUnited States
- Department of Structural Biology, Stanford University School of MedicineStanfordUnited States
| | - Sonia S Majri-Morrison
- Department of Molecular and Cellular Physiology, Stanford University School of MedicineStanfordUnited States
- Department of Structural Biology, Stanford University School of MedicineStanfordUnited States
| | | | - Fei Mo
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, NIHBethesdaUnited States
| | - Peng Li
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, NIHBethesdaUnited States
| | - Magdiel Pérez-Cruz
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of MedicineStanfordUnited States
| | - Peggy P Ho
- Department of Neurology and Neurological Sciences, Stanford UniversityStanfordUnited States
| | - Ievgen Koliesnik
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford UniversityStanfordUnited States
| | - Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford UniversityStanfordUnited States
| | - Tereza Hnizdilova
- Laboratory of Tumor Immunology, Institute of Microbiology of Czech Academy of SciencesPragueCzech Republic
| | - Lora K Picton
- Department of Molecular and Cellular Physiology, Stanford University School of MedicineStanfordUnited States
- Department of Structural Biology, Stanford University School of MedicineStanfordUnited States
| | - Marek Kovar
- Laboratory of Tumor Immunology, Institute of Microbiology of Czech Academy of SciencesPragueCzech Republic
| | - Paul Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford UniversityStanfordUnited States
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford UniversityStanfordUnited States
- Department of Pediatrics, Stanford UniversityStanfordUnited States
| | - Everett Meyer
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of MedicineStanfordUnited States
| | - Jacob Piehler
- Department of Biology, University of OsnabrückOsnabrückGermany
| | - Warren J Leonard
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, NIHBethesdaUnited States
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of MedicineStanfordUnited States
- Department of Structural Biology, Stanford University School of MedicineStanfordUnited States
- Howard Hughes Medical Institute, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
232
|
Cyclic mimetics of kinase-inhibitory region of Suppressors of Cytokine Signaling 1: Progress toward novel anti-inflammatory therapeutics. Eur J Med Chem 2021; 221:113547. [PMID: 34023736 DOI: 10.1016/j.ejmech.2021.113547] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/28/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023]
Abstract
Herein we investigated the structural and cellular effects ensuing from the cyclization of a potent inhibitor of JAK2 as mimetic of SOCS1 protein, named PS5. The introduction of un-natural residues and a lactam internal bridge, within SOCS1-KIR motif, produced candidates that showed high affinity toward JAK2 catalytic domain. By combining CD, NMR and computational studies, we obtained valuable models of the interactions of two peptidomimetics of SOCS1 to deepen their functional behaviors. Notably, when assayed for their biological cell responses mimicking SOCS1 activity, the internal cyclic PS5 analogues demonstrated able to inhibit JAK-mediated tyrosine phosphorylation of STAT1 and to reduce cytokine-induced proinflammatory gene expression, oxidative stress generation and cell migration. The present study well inserts in the field of low-molecular-weight proteomimetics with improved longtime cellular effects and adds a new piece to the puzzled way for the conversion of bioactive peptides into drugs.
Collapse
|
233
|
Weissinger SE, Zahn M, Marienfeld R, Tessmer C, Moldenhauer G, Möller P. A new reliable and highly specific monoclonal antibody to detect the C-terminal region of silencer of cytokine signaling 1. Eur J Haematol 2021; 107:74-80. [PMID: 33714214 DOI: 10.1111/ejh.13620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION SOCS1, a negative regulator of JAK/STAT signaling, is among the most frequently mutated genes in DLBCL and classical Hodgkin lymphoma. The C-terminal SOCS box domain, mediating the degradation of phospho-JAK2, is often affected or even lacking. The analysis of such variants is hampered by the lack of a SOCS1-specific monoclonal antibody recognizing the C-terminus of SOCS1. As this C-terminus is often lost or mutated in B-cell lymphomas, staining with amino-terminal targeting antibodies in a lymphoma setting might be misleading. METHODS BALB/c mice were immunized with a truncated SOCS1 C-terminal protein. The supernatant of generated hybridoma cells was screened by ELISA and, immunohistochemically, on formalin-fixed and paraffin-embedded tonsil. After antibody purification by affinity chromatography, epitope mapping and cross-reactivity check followed via substitution scans. SOCS1 protein expression was investigated on cell cultures and cytoblocks of SOCS1WT stably transfected HEK293T cells, lymphoma cell lines and lymphoid tissues. RESULTS Procedures resulted in one monoclonal IgG1 anti-SOCS1 antibody, 424C, that recognizes and strongly binds to the C-terminal region of SOCS1 in immunoblot and immunohistochemistry analyses. CONCLUSION This new anti-SOCS1 monoclonal antibody is a valuable tool to detect SOCS1 expression dependent on an existing SOCS1 box and, therefore, indicating a full-length SOCS1 protein.
Collapse
Affiliation(s)
| | - Malena Zahn
- Institute of Pathology, University Hospital Ulm, Ulm, Germany
| | - Ralf Marienfeld
- Institute of Pathology, University Hospital Ulm, Ulm, Germany
| | - Claudia Tessmer
- Antibody Unit, Genomics and Proteomics Core Facilities, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gerhard Moldenhauer
- Antibody Unit, Genomics and Proteomics Core Facilities, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Möller
- Institute of Pathology, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
234
|
Stepwise Reversal of Immune Dysregulation Due to STAT1 Gain-of-Function Mutation Following Ruxolitinib Bridge Therapy and Transplantation. J Clin Immunol 2021. [PMID: 33475942 DOI: 10.1007/s10875-020-00943-y/published] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
PURPOSE Patients with heterozygous gain-of-function (GOF) mutations in STAT1 frequently exhibit chronic mucocutaneous candidiasis (CMC), immunodeficiency and autoimmune manifestations. Several treatment options including targeted therapies and hematopoietic stem cell transplantation (HSCT) are available for STAT1 GOF patients but modalities and outcomes are not well established. Herein, we aimed to unravel the effect of ruxolitinib as a bridge therapy in a patient with sporadic STAT1 T385M mutation to manage infections and other disease manifestations. METHODS Peripheral blood mononuclear cells were isolated from the patient prior to, during ruxolitinib treatment and 6 months after HSCT. IFN-β-induced STAT1 phosphorylation/dephosphorylation levels and PMA/ionomycin-stimulated intracellular IL-17A/IFN-γ production in CD4+ T cells were evaluated. Differentially expressed genes between healthy controls and the patient prior to, during ruxolitinib treatment and post-transplantation were investigated using Nanostring nCounter Profiling Panel. RESULTS Ruxolitinib provided favorable responses by controlling candidiasis and autoimmune hemolytic anemia in the patient. Dysregulation in STAT1 phosphorylation kinetics improved with ruxolitinib treatment and was completely normalized after transplantation. TH17 deficiency persisted after ruxolitinib treatment, but normalized following HSCT. Consistent with the impairment in JAK/STAT signaling, multiple immune related pathways were found to be dysregulated in the patient. At baseline, genes related to type I IFN-related pathways, antigen processing, T-cell and B-cell functions were upregulated, while NK-cell function and cytotoxicity related genes were downregulated. Dysregulated gene expression was partially improved with ruxolitinib treatment and normalized after transplantation. CONCLUSION Our findings suggest that improved disease management and immune dysregulatory profile can be achieved with ruxolitinib treatment before transplantation and this would be beneficial to reduce the risk of adverse outcome of HSCT.
Collapse
|
235
|
Ross DM, Babon JJ, Tvorogov D, Thomas D. Persistence of myelofibrosis treated with ruxolitinib: biology and clinical implications. Haematologica 2021; 106:1244-1253. [PMID: 33472356 PMCID: PMC8094080 DOI: 10.3324/haematol.2020.262691] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Indexed: 12/18/2022] Open
Abstract
Activation of JAK-STAT signaling is one of the hallmarks of myelofibrosis, a myeloproliferative neoplasm that leads to inflammation, progressive bone marrow failure, and a risk of leukemic transformation. Around 90% of patients with myelofibrosis have a mutation in JAK2, MPL, or CALR: so-called 'driver' mutations that lead to activation of JAK2. Ruxolitinib, and other JAK2 inhibitors in clinical use, provide clinical benefit but do not have a major impact on the abnormal hematopoietic clone. This phenomenon is termed 'persistence', in contrast to usual patterns of resistance. Multiple groups have shown that type 1 inhibitors of JAK2, which bind the active conformation of the enzyme, lead to JAK2 becoming resistant to degradation with consequent accumulation of phospho-JAK2. In turn, this can lead to exacerbation of inflammatory manifestations when the JAK inhibitor is discontinued, and it may also contribute to disease persistence. The ways in which JAK2 V617F and CALR mutations lead to activation of JAK-STAT signaling are incompletely understood. We summarize what is known about pathological JAK-STAT activation in myelofibrosis and how this might lead to future novel therapies for myelofibrosis with greater disease-modifying potential.
Collapse
Affiliation(s)
- David M Ross
- Department of Hematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide; Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide; Precision Medicine Theme, South Australian Health and Medical Research Institute, and Adelaide Medical School, University of Adelaide.
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, University of Melbourne, Parkville
| | - Denis Tvorogov
- Centre for Cancer Biology, University of South Australia and SA Pathology
| | - Daniel Thomas
- Precision Medicine Theme, South Australian Health and Medical Research Institute, and Adelaide Medical School, University of Adelaide
| |
Collapse
|
236
|
Cellular and Molecular Mechanisms of Breast Implant-Associated Anaplastic Large Cell Lymphoma. Plast Reconstr Surg 2021; 147:30e-41e. [PMID: 33370049 DOI: 10.1097/prs.0000000000007423] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
SUMMARY Breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) is an emerging and highly treatable cancer of the immune system that can form around textured-surface breast implants. Although the underlying cause has yet to be elucidated, an emerging theme-linking pathogenesis to a chronic inflammatory state-continues to dominate the current literature. Specifically, the combination of increasing mutation burden and chronic inflammation leads to aberrant T-cell clonal expansion. However, the impetus remains largely unknown. Proposed mechanisms include a lipopolysaccharide endotoxin response, oncogenic transformation related to viral infection, associated trauma to the breast pocket, particulate matter digestion by capsular macrophages, chronic allergic inflammation, and genetic susceptibility. The Janus kinase-signal transducer and activator of transcription 3 (JAK-STAT3) pathway is a major signaling pathway that regulates a variety of intracellular growth and survival processes. Constitutive activation of JAK-STAT3 has been implicated in several malignancies, including lymphomas, and has recently been identified as a potential key mediator in BIA-ALCL. The purpose of this article is to review the cellular and molecular mechanisms of BIA-ALCL with a focus on the role of oncogenic JAK-STAT3 signaling in BIA-ALCL tumorigenesis and progression. Selected experimental work from the authors' group on aberrant JAK-STAT3 signaling in BIA-ALCL is also included. The authors discuss how an inflammatory microenvironment may facilitate malignant transformation through the JAK-STAT3 pathway-highlighting its potential mechanistic role. The authors' hope is that further investigation of this signaling pathway will reveal avenues for using JAK-STAT3 signaling as a prognostic indicator and novel therapeutic target in the case of advanced disease.
Collapse
|
237
|
Liu C, Zhao Q, Zhong L, Li Q, Li R, Li S, Li Y, Li N, Su J, Dhondrup W, Meng X, Zhang Y, Tu Y, Wang X. Tibetan medicine Ershiwuwei Lvxue Pill attenuates collagen-induced arthritis via inhibition of JAK2/STAT3 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113820. [PMID: 33465441 DOI: 10.1016/j.jep.2021.113820] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/18/2020] [Accepted: 01/11/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ershiwuwei Lvxue Pill (ELP, མགྲིན་མཚལ་ཉེར་ལྔ།), a traditional Tibetan medicine preparation, has been used hundreds of years for the clinical treatment of rheumatoid arthritis (RA) in the highland region of Tibet, China. However, the underlying mechanism of its therapeutic effect remains unclear. AIM OF THE STUDY The present study aimed to investigate the potential pharmacological mechanisms of anti-arthritic effect of ELP. MATERIALS AND METHODS The main chemical constituents of ELP were analyzed by ultra-performance liquid chromatography quadrupole-time-flight mass spectrometry (UPLC-Q-TOF-MS). Forty-eight male Wistar rats (220 ± 20 g) were randomly divided into six groups: normal group, collagen-induced arthritis (CIA) group, methotrexate group (1.05 mg/kg), ELP groups (115, 230 and 460 mg/kg). CIA rat models were assigned to evaluate the anti-RA activity of ELP by determining the paws swelling, arthritis score, organ coefficients of spleen and thymus, and histopathological analysis of knee joints of synovial tissues. The levels of TNF-α, IL-10, IL-6 and IL-17 in serum were measured by ELISA. In addition, mRNA and protein expression levels associated with JAK2/STAT3 signaling pathway in synovial tissues of CIA rats were detected by qRT-PCR, immunohistochemistry and Western blot analyses. RESULTS Fourteen main chemical constituents of ELP were quantitatively determined by UPLC-Q-TOF-MS analysis. Treatment with ELP reduced the paw swelling, arthritis score and organ coefficients of spleen and thymus. Histopathological examination revealed the protective effects of ELP on CIA rats with knee joint injury. The levels of serum pro-inflammatory cytokines (TNF-α, IL-6 and IL-17) were markedly reduced while the anti-inflammatory cytokine IL-10 was significantly increased with the treatment of ELP. Further investigations showed ELP down-regulated the mRNA and protein expression levels of Bcl-2, whereas up-regulated Bax, SOCS1 and SOCS3. Meanwhile, the ratios of p-JAK2/JAK2 and p-STAT3/STAT3 proteins from synovial tissues were dramatically decreased with the treatment of ELP, whereas no changes of the mRNA and protein expression levels of JAK2 and STAT3 were observed. CONCLUSION These results indicated that ELP reduced the severity of arthritis and joint swelling, suggesting an antirheumatic effect on CIA rats. The possible mechanism is related to inhibiting inflammatory response and inducing apoptosis in synovial tissues by regulating JAK2/STAT3 signaling pathway. However, further in vivo and in vitro investigations are still needed to clarify the underlying mechanism of ELP in treating RA.
Collapse
Affiliation(s)
- Chuan Liu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qian Zhao
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lu Zhong
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qiuyue Li
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Rui Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shuang Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yangxin Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ning Li
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jinsong Su
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wüntrang Dhondrup
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianli Meng
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yi Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Ya Tu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Development Research Center of Traditional Chinese Medicine, China Academy of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Xiaobo Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
238
|
Budroni V, Versteeg GA. Negative Regulation of the Innate Immune Response through Proteasomal Degradation and Deubiquitination. Viruses 2021; 13:584. [PMID: 33808506 PMCID: PMC8066222 DOI: 10.3390/v13040584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 12/25/2022] Open
Abstract
The rapid and dynamic activation of the innate immune system is achieved through complex signaling networks regulated by post-translational modifications modulating the subcellular localization, activity, and abundance of signaling molecules. Many constitutively expressed signaling molecules are present in the cell in inactive forms, and become functionally activated once they are modified with ubiquitin, and, in turn, inactivated by removal of the same post-translational mark. Moreover, upon infection resolution a rapid remodeling of the proteome needs to occur, ensuring the removal of induced response proteins to prevent hyperactivation. This review discusses the current knowledge on the negative regulation of innate immune signaling pathways by deubiquitinating enzymes, and through degradative ubiquitination. It focusses on spatiotemporal regulation of deubiquitinase and E3 ligase activities, mechanisms for re-establishing proteostasis, and degradation through immune-specific feedback mechanisms vs. general protein quality control pathways.
Collapse
Affiliation(s)
| | - Gijs A. Versteeg
- Max Perutz Labs, Department of Microbiology, Immunobiology, and Genetics, University of Vienna, Vienna Biocenter (VBC), 1030 Vienna, Austria;
| |
Collapse
|
239
|
Megalocytivirus Induces Complicated Fish Immune Response at Multiple RNA Levels Involving mRNA, miRNA, and circRNA. Int J Mol Sci 2021; 22:ijms22063156. [PMID: 33808870 PMCID: PMC8003733 DOI: 10.3390/ijms22063156] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 12/22/2022] Open
Abstract
Megalocytivirus is an important viral pathogen to many farmed fishes, including Japanese flounder (Paralichthys olivaceus). In this study, we examined megalocytivirus-induced RNA responses in the spleen of flounder by high-throughput sequencing and integrative analysis of various RNA-seq data. A total of 1327 microRNAs (miRNAs), including 368 novel miRNAs, were identified, among which, 171 (named DEmiRs) exhibited significantly differential expressions during viral infection in a time-dependent manner. For these DEmiRs, 805 differentially expressed target mRNAs (DETmRs) were predicted, whose expressions not only significantly changed after megalocytivirus infection but were also negatively correlated with their paired DEmiRs. Integrative analysis of immune-related DETmRs and their target DEmiRs identified 12 hub DEmiRs, which, together with their corresponding DETmRs, formed an interaction network containing 84 pairs of DEmiR and DETmR. In addition to DETmRs, 19 DEmiRs were also found to regulate six key immune genes (mRNAs) differentially expressed during megalocytivirus infection, and together they formed a network consisting of 21 interactive miRNA-messenger RNA (mRNA) pairs. Further analysis identified 9434 circular RNAs (circRNAs), 169 of which (named DEcircRs) showed time-specific and significantly altered expressions during megalocytivirus infection. Integrated analysis of the DETmR-DEmiR and DEcircR-DEmiR interactions led to the identification of a group of competing endogenous RNAs (ceRNAs) constituted by interacting triplets of circRNA, miRNA, and mRNA involved in antiviral immunity. Together these results indicate that complicated regulatory networks of different types of non-coding RNAs and coding RNAs are involved in megalocytivirus infection.
Collapse
|
240
|
de Oliveira LF, de Andrade AAS, Pagliari C, de Carvalho LV, Silveira TS, Cardoso JF, Silva ALTE, de Vasconcelos JM, Moreira-Nunes CA, Burbano RMR, Nunes MRT, Dos Santos EJM, Júnior JLDSGV. Differential expression analysis and profiling of hepatic miRNA and isomiRNA in dengue hemorrhagic fever. Sci Rep 2021; 11:5554. [PMID: 33692368 PMCID: PMC7946910 DOI: 10.1038/s41598-020-72892-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 09/03/2020] [Indexed: 12/13/2022] Open
Abstract
Dengue virus causes dengue hemorrhagic fever (DHF) and has been associated to fatal cases worldwide. The liver is one of the most important target tissues in severe cases, due to its intense viral replication and metabolic role. microRNAs role during infection is crucial to understand the regulatory mechanisms of DENV infection and can help in diagnostic and anti-viral therapies development. We sequenced the miRNome of six fatal cases and compared to five controls, to characterize the human microRNAs expression profile in the liver tissue during DHF. Eight microRNAs were differentially expressed, including miR-126-5p, a regulatory molecule of endothelial cells, miR-122-5p, a liver specific homeostasis regulator, and miR-146a-5p, an interferon-regulator. Enrichment analysis with predicted target genes of microRNAs revealed regulatory pathways of apoptosis, involving MAPK, RAS, CDK and FAS. Immune response pathways were related to NF- kB, CC and CX families, IL and TLR. This is the first description of the human microRNA and isomicroRNA profile in liver tissues from DHF cases. The results demonstrated the association of miR-126-5p, miR-122-5p and miR-146a-5p with DHF liver pathogenesis, involving endothelial repair and vascular permeability regulation, control of homeostasis and expression of inflammatory cytokines.
Collapse
Affiliation(s)
- Layanna Freitas de Oliveira
- Center for Technological Innovation, Instituto Evandro Chagas, Ananindeua, PA, Brazil. .,Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil.
| | | | - Carla Pagliari
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - Taiana S Silveira
- Faculdade de Medicina de São José Do Rio Preto, São Paulo, SP, Brazil
| | | | | | | | - Caroline Aquino Moreira-Nunes
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Universidade Federal Do Ceará, Fortaleza, CE, Brazil
| | | | | | | | | |
Collapse
|
241
|
Cecil D, Park KH, Curtis B, Corulli L, Disis MN. Type I T cells sensitize treatment refractory tumors to chemotherapy through inhibition of oncogenic signaling pathways. J Immunother Cancer 2021; 9:e002355. [PMID: 33762321 PMCID: PMC7993179 DOI: 10.1136/jitc-2021-002355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The most common clinical outcome observed after treatment with immune checkpoint inhibitor antibodies is disease stabilization. Using vaccines to generate high levels of tumor antigen-specific T-helper 1 (Th1), we show that tumors not eradicated by vaccination demonstrate prolonged disease stabilization. We evaluated the mechanism by which type I T cells inhibit disease progression and potentially influence the subsequent clinical response to standard therapy in treatment refractory cancers. METHODS We employed a meta-analysis of studies with tumor growth from four different vaccines in two different mammary cancer models. The T-cell subtype and cytokine essential for vaccine-induced tumor inhibition was determined by in vivo neutralization studies and immunohistochemistry. The role of interferon gamma (IFN-γ) in receptor tyrosine kinase and downstream signaling was determined by immunoblotting. The role of suppressor of cytokine signaling 1 (SOCS1) on IFN-γ signaling was evaluated on SOCS1-silenced cells with immunoblotting and immunoprecipitation. The effect of vaccination on growth factor receptor signaling pathways, performed in both luminal (TgMMTVneu) and basal (C3(1)-Tag) mammary cancer models treated with paclitaxel or an anti-HER2-neu monoclonal antibody were assessed via immunoblotting. RESULTS Immunization with an epitope-based vaccine targeting a representative tumor antigen resulted in elevated tumor trafficking Tbet+CD4 T cells, decreased tumor proliferation and increased apoptosis compared with control vaccinated mice. The resulting disease stabilization was dependent on IFN-γ-secreting CD4+ T cells. In the presence of excess IFN-γ, SOCS1 became upregulated in tumor cells, bound insulin receptor, insulin like growth factor receptor 1 and epidermal growth factor receptor resulting in profound oncogenic signaling inhibition. Silencing SOCS1 restored growth factor receptor signaling and proliferation and prevented cell death. Similar signaling perturbations were detected in vaccinated mice developing antigen-specific Th1 cells. Vaccination synergized with standard therapies and restored disease sensitivity to treatment with both a neu-specific antibody and paclitaxel in TgMMTVneu and to paclitaxel in C3(1)-Tag. Combination of vaccination and chemotherapy or biological therapy was more effective than monotherapy alone in either model and resulted in complete resolution of disease in some individuals. CONCLUSIONS These data suggest the clinical activity of type I T cells extends beyond direct tumor killing and immune therapies designed to increase type I T cells and could be integrated into standard chemotherapy regimens to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Denise Cecil
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| | | | - Benjamin Curtis
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| | - Lauren Corulli
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| | - Mary Nora Disis
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
242
|
Klopfenstein N, Brandt SL, Castellanos S, Gunzer M, Blackman A, Serezani CH. SOCS-1 inhibition of type I interferon restrains Staphylococcus aureus skin host defense. PLoS Pathog 2021; 17:e1009387. [PMID: 33690673 PMCID: PMC7984627 DOI: 10.1371/journal.ppat.1009387] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/22/2021] [Accepted: 02/16/2021] [Indexed: 12/19/2022] Open
Abstract
The skin innate immune response to methicillin-resistant Staphylococcus aureus (MRSA) culminates in the formation of an abscess to prevent bacterial spread and tissue damage. Pathogen recognition receptors (PRRs) dictate the balance between microbial control and injury. Therefore, intracellular brakes are of fundamental importance to tune the appropriate host defense while inducing resolution. The intracellular inhibitor suppressor of cytokine signaling 1 (SOCS-1), a known JAK/STAT inhibitor, prevents the expression and actions of PRR adaptors and downstream effectors. Whether SOCS-1 is a molecular component of skin host defense remains to be determined. We hypothesized that SOCS-1 decreases type I interferon production and IFNAR-mediated antimicrobial effector functions, limiting the inflammatory response during skin infection. Our data show that MRSA skin infection enhances SOCS-1 expression, and both SOCS-1 inhibitor peptide-treated and myeloid-specific SOCS-1 deficient mice display decreased lesion size, bacterial loads, and increased abscess thickness when compared to wild-type mice treated with the scrambled peptide control. SOCS-1 deletion/inhibition increases phagocytosis and bacterial killing, dependent on nitric oxide release. SOCS-1 inhibition also increases the levels of type I and type II interferon levels in vivo. IFNAR deletion and antibody blockage abolished the beneficial effects of SOCS-1 inhibition in vivo. Notably, we unveiled that hyperglycemia triggers aberrant SOCS-1 expression that correlates with decreased overall IFN signatures in the infected skin. SOCS-1 inhibition restores skin host defense in the highly susceptible hyperglycemic mice. Overall, these data demonstrate a role for SOCS-1-mediated type I interferon actions in host defense and inflammation during MRSA skin infection.
Collapse
Affiliation(s)
- Nathan Klopfenstein
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Stephanie L Brandt
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Sydney Castellanos
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Hufelandstrasse Essen, Germany
- Leibniz-Institut für Analytische Wissenschaften-ISAS -e.V, Dortmund, Germany
| | - Amondrea Blackman
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - C Henrique Serezani
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
243
|
SILAC proteomics implicates SOCS1 in modulating cellular macromolecular complexes and the ubiquitin conjugating enzyme UBE2D involved in MET receptor tyrosine kinase downregulation. Biochimie 2021; 182:185-196. [PMID: 33493533 DOI: 10.1016/j.biochi.2021.01.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/27/2020] [Accepted: 01/18/2021] [Indexed: 01/25/2023]
Abstract
Suppressor of Cytokine Signaling 1 (SOCS1) functions as a tumor suppressor in hepatocellular carcinoma and many other types of cancers. SOCS1 mediates its functions by inhibiting tyrosine kinases, promoting ubiquitination and proteasomal degradation of signal transducing proteins, and by modulating transcription factors. Here, we studied the impact of SOCS1 on the hepatocyte proteome using Stable Isotopic Labelling of Amino acids in Cell culture (SILAC)-based mass spectrometry on the Hepa1-6 murine HCC cell line stably expressing wildtype SOCS1 or a mutant SOCS1 with impaired SH2 domain. As SOCS1 regulates the hepatocyte growth factor (HGF) receptor, the MET receptor tyrosine kinase (RTK), the SILAC-labelled cells were stimulated or not with HGF. Following mass spectrometry analysis, differentially modulated proteins were identified, quantified and analyzed for pathway enrichment. Of the 3440 proteins identified in Hepa-SOCS1 cells at steady state, 181 proteins were significantly modulated compared to control cells. The SH2 domain mutation and HGF increased the number of differentially modulated proteins. Protein interaction network analysis revealed enrichment of SOCS1-modulated proteins within multiprotein complexes such as ubiquitin conjugating enzymes, proteasome, mRNA spliceosome, mRNA exosome and mitochondrial ribosome. Notably, the expression of UBE2D ubiquitin conjugating enzyme, which is implicated in the control of growth factor receptor tyrosine kinase signaling, was found to be regulated by SOCS1. These findings suggest that SOCS1, induced by cytokines, growth factors and diverse other stimuli, has the potential to dynamically modulate of large macromolecular regulatory complexes to help maintain cellular homeostasis.
Collapse
|
244
|
Stepwise Reversal of Immune Dysregulation Due to STAT1 Gain-of-Function Mutation Following Ruxolitinib Bridge Therapy and Transplantation. J Clin Immunol 2021; 41:769-779. [PMID: 33475942 DOI: 10.1007/s10875-020-00943-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/07/2020] [Indexed: 01/09/2023]
Abstract
PURPOSE Patients with heterozygous gain-of-function (GOF) mutations in STAT1 frequently exhibit chronic mucocutaneous candidiasis (CMC), immunodeficiency and autoimmune manifestations. Several treatment options including targeted therapies and hematopoietic stem cell transplantation (HSCT) are available for STAT1 GOF patients but modalities and outcomes are not well established. Herein, we aimed to unravel the effect of ruxolitinib as a bridge therapy in a patient with sporadic STAT1 T385M mutation to manage infections and other disease manifestations. METHODS Peripheral blood mononuclear cells were isolated from the patient prior to, during ruxolitinib treatment and 6 months after HSCT. IFN-β-induced STAT1 phosphorylation/dephosphorylation levels and PMA/ionomycin-stimulated intracellular IL-17A/IFN-γ production in CD4+ T cells were evaluated. Differentially expressed genes between healthy controls and the patient prior to, during ruxolitinib treatment and post-transplantation were investigated using Nanostring nCounter Profiling Panel. RESULTS Ruxolitinib provided favorable responses by controlling candidiasis and autoimmune hemolytic anemia in the patient. Dysregulation in STAT1 phosphorylation kinetics improved with ruxolitinib treatment and was completely normalized after transplantation. TH17 deficiency persisted after ruxolitinib treatment, but normalized following HSCT. Consistent with the impairment in JAK/STAT signaling, multiple immune related pathways were found to be dysregulated in the patient. At baseline, genes related to type I IFN-related pathways, antigen processing, T-cell and B-cell functions were upregulated, while NK-cell function and cytotoxicity related genes were downregulated. Dysregulated gene expression was partially improved with ruxolitinib treatment and normalized after transplantation. CONCLUSION Our findings suggest that improved disease management and immune dysregulatory profile can be achieved with ruxolitinib treatment before transplantation and this would be beneficial to reduce the risk of adverse outcome of HSCT.
Collapse
|
245
|
Chhabra Y, Lee CMM, Müller AF, Brooks AJ. GHR signalling: Receptor activation and degradation mechanisms. Mol Cell Endocrinol 2021; 520:111075. [PMID: 33181235 DOI: 10.1016/j.mce.2020.111075] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022]
Abstract
Growth hormone (GH) actions via initiating cell signalling through the GH receptor (GHR) are important for many physiological processes, in addition to its well-known role in regulating growth. The activation of JAK-STAT signalling by GH is well characterized, however knowledge on GH activation of SRC family kinases (SFKs) is still limited. In this review we summarise the collective knowledge on the activation, regulation, and downstream signalling of GHR. We highlight studies on GH activation of SFKs and the important outcome of this signalling pathway with a focus on the different degradation mechanisms that can regulate GHR availability since this is an area that warrants further study considering its role in tumour progression.
Collapse
Affiliation(s)
- Yash Chhabra
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia; Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21231, USA
| | - Christine M M Lee
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Alexandra Franziska Müller
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Andrew J Brooks
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
246
|
García-Baos A, Alegre-Zurano L, Cantacorps L, Martín-Sánchez A, Valverde O. Role of cannabinoids in alcohol-induced neuroinflammation. Prog Neuropsychopharmacol Biol Psychiatry 2021; 104:110054. [PMID: 32758518 DOI: 10.1016/j.pnpbp.2020.110054] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/13/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
Alcohol is a psychoactive substance highly used worldwide, whose harmful use might cause a broad range of mental and behavioural disorders. Underlying brain impact, the neuroinflammatory response induced by alcohol is recognised as a key contributing factor in the progression of other neuropathological processes, such as neurodegeneration. These sequels are determined by multiple factors, including age of exposure. Strikingly, it seems that the endocannabinoid system modulation could regulate the alcohol-induced neuroinflammation. Although direct CB1 activation can worsen alcohol consequences, targeting other components of the expanded endocannabinoid system may counterbalance the pro-inflammatory response. Indeed, specific modulations of the expanded endocannabinoid system have been proved to exert anti-inflammatory effects, primarily through the CB2 and PPARγ signalling. Among them, some endo- and exogeneous cannabinoids can block certain pro-inflammatory mediators, such as NF-κB, thereby neutralizing the neuroinflammatory intracellular cascades. Furthermore, a number of cannabinoids are able to activate complementary anti-inflammatory pathways, which are necessary for the transition from chronically overactivated microglia to a regenerative microglial phenotype. Thus, cannabinoid modulation provides cooperative anti-inflammatory mechanisms that may be advantageous to resolve a pathological neuroinflammation in an alcohol-dependent context.
Collapse
Affiliation(s)
- Alba García-Baos
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Laia Alegre-Zurano
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Lídia Cantacorps
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ana Martín-Sánchez
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Programme, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Programme, IMIM-Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
247
|
YOSHIMURA A, AKI D, ITO M. SOCS, SPRED, and NR4a: Negative regulators of cytokine signaling and transcription in immune tolerance. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2021; 97:277-291. [PMID: 34121041 PMCID: PMC8403526 DOI: 10.2183/pjab.97.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cytokines are important intercellular communication tools for immunity. Most cytokines utilize the JAK-STAT and Ras-ERK pathways to promote gene transcription and proliferation; however, this signaling is tightly regulated. The suppressor of cytokine signaling (SOCS) family and SPRED family are a representative negative regulators of the JAK-STAT pathway and the Ras-ERK pathway, respectively. The SOCS family regulates the differentiation and function of CD4+ T cells, CD8+ T cells, and regulatory T cells, and is involved in immune tolerance, anergy, and exhaustion. SPRED family proteins have been shown to inactivate Ras by recruiting the Ras-GTPase neurofibromatosis type 1 (NF1) protein. Human genetic analysis has shown that SOCS family members are strongly associated with autoimmune diseases, allergies, and tumorigenesis, and SPRED1 is involved in NF1-like syndromes and tumors. We also identified the NR4a family of nuclear receptors as a key transcription factor for immune tolerance that suppresses cytokine expression and induces various immuno-regulatory molecules including SOCS1.
Collapse
Affiliation(s)
- Akihiko YOSHIMURA
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Correspondence should be addressed: A. Yoshimura, Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan (e-mail: )
| | - Daisuke AKI
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Minako ITO
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
248
|
Leeman-Neill RJ, Soderquist CR, Montanari F, Raciti P, Park D, Radeski D, Mansukhani MM, Murty VV, Hsiao S, Alobeid B, Bhagat G. Phenogenomic heterogeneity of post-transplant plasmablastic lymphomas. Haematologica 2020; 107:201-210. [PMID: 33297669 PMCID: PMC8719101 DOI: 10.3324/haematol.2020.267294] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Indexed: 11/14/2022] Open
Abstract
Plasmablastic lymphoma (PBL) is a rare and clinically aggressive neoplasm that typically occurs in immunocompromised individuals, including those infected with human immunodeficiency virus (HIV) and solid organ allograft recipients. Most prior studies have focused on delineating the clinico-pathological features and genetic attributes of HIVrelated PBL, in which MYC deregulation, Epstein-Barr virus (EBV) infection and, more recently, mutations in JAK/STAT, MAP kinase, and NOTCH pathway genes have been implicated in disease pathogenesis. The phenotypic spectrum of post-transplant (PT)-PBL is not well characterized and data on underlying genetic alterations are limited. This led us to perform comprehensive histopathological and immunophenotypic evaluation and targeted sequencing of 18 samples from 11 patients (8 males, 3 females; age range, 12-76 years) with PT-PBL; eight de novo and three preceded by other types of post-transplant lymphoproliferative disorders. Post-transplant PBL displayed morphological and immunophenotypic heterogeneity and some features overlapped those of plasmablastic myeloma. Six (55%) cases were EBV positive and five (45%) showed MYC rearrangement by fluorescence in situ hybridization. Recurrent mutations in epigenetic regulators (KMT2/MLL family, TET2) and DNA damage repair and response (TP53, mismatch repair genes, FANCA, ATRX), MAP kinase (KRAS, NRAS, HRAS, BRAF), JAK/STAT (STAT3, STAT6, SOCS1), NOTCH (NOTCH1, NOTCH3, SPEN), and immune surveillance (FAS, CD58) pathway genes were observed, with the mutational profiles of EBV+ and EBV– cases exhibiting both similarities and differences. Clinical outcomes also varied, with survival ranging from 0-15.9 years after diagnosis. Besides uncovering the biological heterogeneity of PT-PBL, our study highlights similarities and distinctions between PT-PBL and PBL occurring in other settings and reveals potentially targetable oncogenic pathways in subsets of the disease.
Collapse
Affiliation(s)
| | | | - Francesca Montanari
- Division of Hematology/Oncology, Columbia University Irving Medical Center, NY Presbyterian Hospital, New York, NY
| | | | | | - Dejan Radeski
- Department of Haematology, Sir Charles Gairdner Hospital, Perth
| | | | - Vundavalli V Murty
- Department of Medicine, Division of Cytogenetics, Columbia University Irving Medical Center, NY Presbyterian Hospital, New York, NY
| | | | | | | |
Collapse
|
249
|
Wang S, Ling Y, Yao Y, Zheng G, Chen W. Luteolin inhibits respiratory syncytial virus replication by regulating the MiR-155/SOCS1/STAT1 signaling pathway. Virol J 2020; 17:187. [PMID: 33239033 PMCID: PMC7688008 DOI: 10.1186/s12985-020-01451-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
Background Respiratory syncytial virus (RSV) is a major cause of acute lower respiratory tract infection in infants, children, immunocompromised adults, and elderly individuals. Currently, there are few therapeutic options available to prevent RSV infection. The present study aimed to investigate the effects of luteolin on RSV replication and the related mechanisms. Material and methods We pretreated cells and mice with luteolin before infection with RSV, the virus titer, expressions of RSV-F, interferon (IFN)-stimulated genes (ISGs), and production of IFN-α and IFN-β were determined by plaque assay, RT-qPCR, and ELISA, respectively. The activation of Janus kinase (JAK)-signal transducer and activator of transcription 1 (STAT1) signaling pathway was detected by Western blotting and luciferase assay. Proteins which negatively regulate STAT1 were determined by Western blotting. Then cells were transfected with suppressor of cytokine signaling 1 (SOCS1) plasmid and virus replication and ISGs expression were determined. Luciferase reporter assay and Western blotting were performed to detect the relationship between SOCS1 and miR-155. Results Luteolin inhibited RSV replication, as shown by the decreased viral titer and RSV-F mRNA expression both in vitro and in vivo. The antiviral activity of luteolin was attributed to the enhanced phosphorylation of STAT1, resulting in the increased production of ISGs. Further study showed that SOCS1 was downregulated by luteolin and SOCS1 is a direct target of microRNA-155 (miR-155). Inhibition of miR-155 rescued luteolin-mediated SOCS1 downregulation, whereas upregulation of miR-155 enhanced the inhibitory effect of luteolin. Conclusion Luteolin inhibits RSV replication by regulating the miR-155/SOCS1/STAT1 signaling pathway.
Collapse
Affiliation(s)
- Saisai Wang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Yiting Ling
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Yuanyuan Yao
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Gang Zheng
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Wenbin Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.
| |
Collapse
|
250
|
Eren E, Planès R, Bagayoko S, Bordignon P, Chaoui K, Hessel A, Santoni K, Pinilla M, Lagrange B, Burlet‐Schiltz O, Howard JC, Henry T, Yamamoto M, Meunier E. Irgm2 and Gate-16 cooperatively dampen Gram-negative bacteria-induced caspase-11 response. EMBO Rep 2020; 21:e50829. [PMID: 33124769 PMCID: PMC7645206 DOI: 10.15252/embr.202050829] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/11/2020] [Accepted: 09/25/2020] [Indexed: 12/20/2022] Open
Abstract
Inflammatory caspase-11 (rodent) and caspases-4/5 (humans) detect the Gram-negative bacterial component LPS within the host cell cytosol, promoting activation of the non-canonical inflammasome. Although non-canonical inflammasome-induced pyroptosis and IL-1-related cytokine release are crucial to mount an efficient immune response against various bacteria, their unrestrained activation drives sepsis. This suggests that cellular components tightly control the threshold level of the non-canonical inflammasome in order to ensure efficient but non-deleterious inflammatory responses. Here, we show that the IFN-inducible protein Irgm2 and the ATG8 family member Gate-16 cooperatively counteract Gram-negative bacteria-induced non-canonical inflammasome activation, both in cultured macrophages and in vivo. Specifically, the Irgm2/Gate-16 axis dampens caspase-11 targeting to intracellular bacteria, which lowers caspase-11-mediated pyroptosis and cytokine release. Deficiency in Irgm2 or Gate16 induces both guanylate binding protein (GBP)-dependent and GBP-independent routes for caspase-11 targeting to intracellular bacteria. Our findings identify molecular effectors that fine-tune bacteria-activated non-canonical inflammasome responses and shed light on the understanding of the immune pathways they control.
Collapse
Affiliation(s)
- Elif Eren
- Institute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
| | - Rémi Planès
- Institute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
| | - Salimata Bagayoko
- Institute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
| | - Pierre‐Jean Bordignon
- Institute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
| | - Karima Chaoui
- Institute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
- Mass Spectrometry Core FacilityInstitute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
| | - Audrey Hessel
- Institute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
| | - Karin Santoni
- Institute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
| | - Miriam Pinilla
- Institute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
| | - Brice Lagrange
- CIRI, Centre International de Recherche en InfectiologieInserm, U1111CNRS, UMR5308École Normale Supérieure de LyonUniversité Claude Bernard Lyon 1Univ LyonLyonFrance
| | - Odile Burlet‐Schiltz
- Institute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
- Mass Spectrometry Core FacilityInstitute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
| | - Jonathan C Howard
- Fundação Calouste GulbenkianInstituto Gulbenkian de CiênciaOeirasPortugal
| | - Thomas Henry
- CIRI, Centre International de Recherche en InfectiologieInserm, U1111CNRS, UMR5308École Normale Supérieure de LyonUniversité Claude Bernard Lyon 1Univ LyonLyonFrance
| | - Masahiro Yamamoto
- Department of ImmunoparasitologyResearch Institute for Microbial DiseasesOsaka UniversityOsakaJapan
- Laboratory of ImmunoparasitologyWPI Immunology Frontier Research CenterOsaka UniversityOsakaJapan
| | - Etienne Meunier
- Institute of Pharmacology and Structural Biology (IPBS)CNRS, UMR5089University of ToulouseToulouseFrance
- Present address:
Institute of Pharmacology and Structural Biology (IPBS)CNRSToulouseFrance
| |
Collapse
|