1
|
Sapra L, Saini C, Sharma S, Nanda D, Nilakhe A, Chattopadhyay N, Meena AS, Mishra PK, Gupta S, Garg B, Manhas V, Srivastava RK. Targeting the osteoclastogenic cytokine IL-9 as a novel immunotherapeutic strategy in mitigating inflammatory bone loss in post-menopausal osteoporosis. JBMR Plus 2024; 8:ziae120. [PMID: 39399159 PMCID: PMC11470976 DOI: 10.1093/jbmrpl/ziae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/24/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Recent discoveries have established the pivotal role of IL-9-secreting immune cells in a wide spectrum of inflammatory and autoimmune diseases. However, little is known about how IL-9 contributes to the etiology of inflammatory bone loss in PMO. We observed that IL-9 has a pathological impact on inflammatory bone loss in ovariectomized (Ovx) mice. Our in vivo temporal kinetics analysis revealed that estrogen deprivation enhanced the production of IL-9 from Th cells (majorly Th9 and Th17). Both our ex vivo and in vivo studies corroborated these findings in Ovx mice, as estrogen diminishes the potential of Th9 cells to produce IL-9. Mechanistically, Th9 cells in an IL-9-dependent manner enhance osteoclastogenesis and thus could establish themselves as a novel osteoclastogenic Th cell subset. Therapeutically neutralizing/blocking IL-9 improves bone health by inhibiting the differentiation and function of osteoclasts, Th9, and Th17 cells along with maintaining gut integrity in Ovx mice. Post-menopausal osteoporotic patients have increased IL-9-secreting Th9 cells, which may suggest a potential role for IL-9 in the development of osteoporosis. Collectively, our study identifies IL-9-secreting Th9 cells as a driver of bone loss with attendant modulation of gut-immune-bone axis, which implies IL-9-targeted immunotherapies as a potential strategy for the management and treatment of inflammatory bone loss observed in PMO.
Collapse
Affiliation(s)
- Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Chaman Saini
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Shivani Sharma
- Division of Endocrinology, Central Drug Research Institute (CDRI), Lucknow 226031, India
| | - Dibyani Nanda
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | | | - Naibedya Chattopadhyay
- Division of Endocrinology, Central Drug Research Institute (CDRI), Lucknow 226031, India
| | - Avtar Singh Meena
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pradyumna K Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, MP 462001, India
| | - Sarika Gupta
- National Institute of Immunology (NII), New Delhi 110067, India
| | - Bhavuk Garg
- Department of Orthopaedics, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Vikrant Manhas
- Department of Orthopaedics, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| |
Collapse
|
2
|
Chandwaskar R, Dalal R, Gupta S, Sharma A, Parashar D, Kashyap VK, Sohal JS, Tripathi SK. Dysregulation of T cell response in the pathogenesis of inflammatory bowel disease. Scand J Immunol 2024:e13412. [PMID: 39394898 DOI: 10.1111/sji.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024]
Abstract
Inflammatory bowel disease (IBD), comprised of Crohn's disease (CD) and ulcerative colitis (UC), are gut inflammatory diseases that were earlier prevalent in the Western Hemisphere but now are on the rise in the East, with India standing second highest in the incidence rate in the world. Inflammation in IBD is a cause of dysregulated immune response, wherein helper T (Th) cell subsets and their cytokines play a major role in the pathogenesis of IBD. In addition, gut microbiota, environmental factors such as dietary factors and host genetics influence the outcome and severity of IBD. Dysregulation between effector and regulatory T cells drives gut inflammation, as effector T cells like Th1, Th17 and Th9 subsets Th cell lineages were found to be increased in IBD patients. In this review, we attempted to discuss the role of different Th cell subsets together with other T cells like CD8+ T cells, NKT and γδT cells in the outcome of gut inflammation in IBD. We also highlighted the potential therapeutic candidates for IBD.
Collapse
Affiliation(s)
- Rucha Chandwaskar
- Amity Institute of Microbial Technology (AIMT), Amity University Jaipur, Rajasthan, India
| | - Rajdeep Dalal
- Infection and Immunology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, India
| | - Saurabh Gupta
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Aishwarya Sharma
- Sri Siddhartha Medical College and Research Center, Tumkur, Karnataka, India
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vivek K Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Jagdip Singh Sohal
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Subhash K Tripathi
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
3
|
Bick F, Blanchetot C, Lambrecht BN, Schuijs MJ. A reappraisal of IL-9 in inflammation and cancer. Mucosal Immunol 2024:S1933-0219(24)00106-5. [PMID: 39389468 DOI: 10.1016/j.mucimm.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
While much is known about the functional effects of type 2 cytokines interleukin (IL)-4, IL-5 and IL-13 in homeostasis and disease, we still poorly understand the functions of IL-9. Chronic inflammation seen in allergic diseases, autoimmunity and cancer is however frequently accompanied by overproduction of this elusive type 2 cytokine. Initially identified as a T cell and mast cell growth factor, and later as the hallmark cytokine defining TH9 cells, we now know that IL-9 is produced by multiple innate and adaptive immune cells. Recent evidence suggests that IL-9 controls discrete aspects of the allergic cascade, cellular responses of immune and stromal cells, cancer progression, tolerance and immune escape. Despite functioning as a pleiotropic cytokine in mucosal environments, like the lungs, the direct and indirect cellular targets of IL-9 are still not well characterized. Here, we discuss IL-9's cellular senders and receivers, focusing on asthma and cancer. Moreover, we review current research directions and the outlook of targeted therapy centered around the biology of IL-9.
Collapse
Affiliation(s)
- Fabian Bick
- argenx BV, 9052 Zwijnaarde, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium
| | | | - Bart N Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Martijn J Schuijs
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium.
| |
Collapse
|
4
|
Zhang L, Zhu Z, Zheng L, Liu X, Li H, Dai X, Zhang Z, Wang B, Huang X, Ren Q, Xu Y. Identification of a FOXO gene and its roles in anti-WSSV infection through regulation of Dicers and Argos in Macrobrachium nipponense. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109908. [PMID: 39299407 DOI: 10.1016/j.fsi.2024.109908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Forkhead box O (FOXO) proteins are a subgroup of the forkhead family of transcription factors that play important roles in the immune response. In this study, we cloned and identified a FOXO gene named MnFOXO from Macrobrachium nipponense. The full-length cDNA of MnFOXO is 2086 bp and contains a 1302 bp open reading frame, which encodes 433 amino acids. MnFOXO consists of five low-complexity regions and a conserved DNA-binding domain (forkhead domain). Evolutionary analyses indicate that MnFOXO proteins cluster with FOXO proteins from crustaceans. Tissue distribution analysis showed that MnFOXO was expressed in all detected tissues, with relatively higher expression levels in the intestine, eyestalks, stomach, and hemocytes than in the hepatopancreas, gills, and heart. The expression levels of MnFOXO in the hepatopancreas and intestine were significantly up-regulated in M. nipponense infected with white spot syndrome virus (WSSV) at 24 and 48 h. Furthermore, knockdown of MnFOXO increased the expression of WSSV envelope protein VP28 during WSSV infection. Further studies showed that knockdown of the MnFOXO gene in M. nipponense inhibited the synthesis of Dicers (MnDicer1, MnDicer2) and Argonautes (MnArgo1, MnArgo2) during WSSV invasion. These findings suggest that MnFOXO positively regulates the expression of Dicers and Argos, and inhibits the expression of VP28. This study provides new evidence for understanding the role of FOXO in antiviral innate immunity in crustaceans.
Collapse
Affiliation(s)
- Lihua Zhang
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, China
| | - Ziyue Zhu
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, China
| | - Liangmin Zheng
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, China
| | - Xiaohan Liu
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, China
| | - Hao Li
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, China
| | - Xiaoling Dai
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, China
| | - Zhaoqian Zhang
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, China
| | - Bingyan Wang
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, China
| | - Xin Huang
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, China.
| | - Qian Ren
- School of Marine Sciences, Nanjing University of information Science & Technology, Nanjing, Jiangsu Province, 210044, PR China.
| | - Yu Xu
- Key Laboratory of Genetic Breeding and cultivation for Freshwater Crustacean, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing 210017, China.
| |
Collapse
|
5
|
Feng K, Cen J, Zou X, Zhang T. Novel insight into MDA-7/IL-24: A potent therapeutic target for autoimmune and inflammatory diseases. Clin Immunol 2024; 266:110322. [PMID: 39033900 DOI: 10.1016/j.clim.2024.110322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
Melanoma differentiation-associated gene-7 (MDA-7)/interleukin-24 (IL-24) is a pleiotropic member of the IL-10 family of cytokines, and is involved in multiple biological processes, including cell proliferation, cell differentiation, tissue fibrosis, the inflammatory response, and antitumor activity. MDA-7/IL-24 can regulate epithelial integrity, homeostasis, mucosal immunity and host resistance to various pathogens by enhancing immune and inflammatory responses. Our recent study revealed the mechanism of MDA-7/IL-24 in promoting airway inflammation and airway remodeling through activating the JAK/STAT3 and ERK signaling pathways in bronchial epithelial cells. Herein, we summarize the cellular sources, inducers, target cells, signaling pathways, and biological effects of MDA-7/IL-24 in several allergic and autoimmune diseases. This review also synopsizes recent advances in clinical research targeting MDA-7/IL-24 or its receptors. Based on these advancements, we emphasize its potential as a target for immunotherapy and discuss the challenges of developing immunotherapeutic drugs targeting MDA-7/IL-24 or its receptors in autoimmune and inflammatory disorders.
Collapse
Affiliation(s)
- Kangni Feng
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
| | - Jiemei Cen
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
| | - Xiaoling Zou
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
| | - Tiantuo Zhang
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China.
| |
Collapse
|
6
|
Narayan S, Dalal R, Rizvi ZA, Awasthi A. Zinc dampens antitumor immunity by promoting Foxp3 + regulatory T cells. Front Immunol 2024; 15:1389387. [PMID: 39247196 PMCID: PMC11377231 DOI: 10.3389/fimmu.2024.1389387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/26/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction The role of zinc (Zn) in tumor development and immune modulation has always been paradoxical. This study redefines our understanding of the impact of Zn on cancer progression and therapeutic strategies. Methods We investigated the effects of dietary Zn levels on tumor progression and immune responses. This included examining the impact of both high and deficient dietary Zn, as well as Zn chelation, on tumor growth and immune cell populations. Specifically, we analyzed the frequency of Foxp3+ regulatory T-cells (Tregs) and identified the role of FOXO1 in Zn-mediated effects on Tregs. Additionally, we explored the therapeutic potential of clioquinol (CQ) in enhancing α-PD-1 immunotherapy responses, particularly in melanoma. Results Our findings show that high dietary Zn promotes tumor progression by fostering a protumorigenic environment mediated by T cells. Increased Zn intake was found to facilitate tumor progression by increasing Foxp3+ Treg frequency. In contrast, deficiency in dietary Zn and chelation of tissue Zn emerged as potent drivers of antitumor immunity. We pinpointed FOXO1 as the master regulator governing the influence of Zn on Tregs. Discussion These results reveal a novel mechanistic insight into how Zn influences tumor progression and immune regulation. The identification of FOXO1 as a key regulator opens new avenues for understanding the role of Zn in cancer biology. Furthermore, we introduce a promising therapeutic approach by showing that administering clioquinol (CQ) significantly enhances α-PD-1 immunotherapy response, particularly in melanoma. These revelations transform our comprehension of the multifaceted role of Zn in tumorigenesis and immune regulation, highlighting innovative possibilities for cancer therapy.
Collapse
Affiliation(s)
- Sugandha Narayan
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, National Capital Region (NCR)-Biotech Science Cluster, Faridabad, Haryana, India
| | - Rajdeep Dalal
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, National Capital Region (NCR)-Biotech Science Cluster, Faridabad, Haryana, India
| | - Zaigham Abbas Rizvi
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, National Capital Region (NCR)-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology Core Lab, Translational Health Science and Technology Institute, National Capital Region (NCR)-Biotech Science Cluster, Faridabad, Haryana, India
| | - Amit Awasthi
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, National Capital Region (NCR)-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology Core Lab, Translational Health Science and Technology Institute, National Capital Region (NCR)-Biotech Science Cluster, Faridabad, Haryana, India
| |
Collapse
|
7
|
Madan U, Verma B, Awasthi A. Cenicriviroc, a CCR2/CCR5 antagonist, promotes the generation of type 1 regulatory T cells. Eur J Immunol 2024; 54:e2350847. [PMID: 38643381 DOI: 10.1002/eji.202350847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/22/2024]
Abstract
Cenicriviroc, a dual CCR2/CCR5 antagonist, initially developed as an anti-HIV drug, has shown promising results in nonalcoholic steatohepatitis phase 2 clinical trials. It inhibits the infiltration and activation of CCR2+/CCR5+ monocytes and macrophages to the site of liver injury, preventing liver fibrosis. However, the role of Cenicriviroc in the modulation of helper T cell differentiation and functions remains to be explored. In inflamed colons of Crohn's disease patients, CCR2+ and CCR5+ CD4+ T cells are enriched. Considering the role of CCR2+ and CCR5+ T cells in IBD pathogenesis, we investigated the potential role of Cenicriviroc in colitis. Our in vitro studies revealed that Cenicriviroc inhibits Th1-, Th2-, and Th17-cell differentiation while promoting the generation of type 1 regulatory T cells (Tr1), known for preventing inflammation through induction of IL-10. This study is the first to report that Cenicriviroc promotes Tr1 cell generation by up-regulating the signature of Tr1 cell transcription factors such as c-Maf, Prdm1, Irf-1, Batf, and EGR-2. Cenicriviroc displayed a protective effect in experimental colitis models by preventing body weight loss and intestinal inflammation and preserving epithelial barrier integrity. We show that Cenicriviroc induced IL-10 and inhibited the generation of pro-inflammatory cytokines IFN-γ, IL-17, IL-6, and IL-1β during colitis. Based on our data, we propose Cenicriviroc as a potential therapeutic in controlling tissue inflammation by inhibiting the generation and functions of effector T cells and promoting the induction of anti-inflammatory Tr1 cells.
Collapse
Affiliation(s)
- Upasna Madan
- Centre for Immuno-biology and Immunotherapy, NCR-Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Bhawna Verma
- Centre for Immuno-biology and Immunotherapy, NCR-Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Amit Awasthi
- Centre for Immuno-biology and Immunotherapy, NCR-Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, NCR Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
8
|
Wang Y, Shang P, Xu C, Dong W, Zhang X, Xia Y, Sui C, Yang C. Novel genetic alterations in liver cancer distinguish distinct clinical outcomes and combination immunotherapy responses. Front Pharmacol 2024; 15:1416295. [PMID: 38948469 PMCID: PMC11211383 DOI: 10.3389/fphar.2024.1416295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction: Genomic profiling has revolutionized therapeutic interventions and the clinical management of liver cancer. However, pathogenetic mechanisms, molecular determinants of recurrence, and predictive biomarkers for first-line treatment (anti-PD-(L)1 plus bevacizumab) in liver cancer remain incompletely understood. Materials and methods: Targeted next-generation sequencing (tNGS) (a 603-cancer-gene panel) was applied for the genomic profiling of 232 hepatocellular carcinoma (HCC) and 22 intrahepatic cholangiocarcinoma (ICC) patients, among which 47 unresectable/metastatic HCC patients underwent anti-PD-1 plus bevacizumab therapy. Genomic alterations were estimated for their association with vascular invasion (VI), location of onset, recurrence, overall survival (OS), recurrence-free survival (RFS), and anti-PD-1 plus bevacizumab therapy response. Results: The genomic landscape exhibited that the most commonly altered genes in HCC were TP53, FAT3, PDE4DIP, KMT2C, FAT1, and MYO18A, while TP53, FAT1, FAT3, PDE4DIP, ROS1, and GALNT11 were frequently altered in ICC; notably, KRAS (18.18% vs. 1.29%) and BAP1 (13.64% vs. 1.29%) alterations were significantly more prevalent in ICC. Comparison analysis demonstrated the distinct clinicopathological/genomic characterizations between Chinese and Western HCC cohorts. Genomic profiling of HCC underlying VI showed that LDLR, MSH2, KDM5D, PDE3A, and FOXO1 were frequently altered in the VI group compared to patients without VIs. Compared to the right hepatic lobes of HCC patients, the left hepatic lobe of HCC patients had superior OS (median OS: 36.77 months vs. unreached, p < 0.05). By further comparison, Notch signaling pathway-related alterations were significantly prevalent among the right hepatic lobes of HCC patients. Of note, multivariate Cox regression analysis showed that altered RB1, NOTCH3, MGA, SYNE1, and ZFHX3, as independent prognostic factors, were significantly correlated with the OS of HCC patients. Furthermore, altered LATS1 was abundantly enriched in the HCC-recurrent group, and impressively, it was independent of clinicopathological features in predicting RFS (median RFS of altered type vs. wild-type: 5.57 months vs. 22.47 months, p < 0.01). Regarding those treated HCC patients, TMB value, altered PTPRZ1, and cell cycle-related alterations were identified to be positively associated with the objective response rate (ORR), but KMT2D alterations were negatively correlated with ORR. In addition, altered KMT2D and cell cycle signaling were significantly associated with reduced and increased time to progression-free survival (PFS), respectively. Conclusion: Comprehensive genomic profiling deciphered distinct molecular characterizations underlying VI, location of onset, recurrence, and survival time in liver cancer. The identification of novel genetic predictors of response to anti-PD-1 plus bevacizumab in HCC facilitated the development of an evidence-based approach to therapy.
Collapse
Affiliation(s)
- Yizhou Wang
- Department of Hepatic Surgery IV and Clinical Research Institute, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Peipei Shang
- Department of Medical Oncology, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chang Xu
- Department of General Surgery, Biliary Tract Disease Institute, Biliary Tract Disease Center, and Cancer Center of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Dong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiaofeng Zhang
- Department of Hepatic Surgery IV and Clinical Research Institute, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yong Xia
- Department of Hepatic Surgery IV and Clinical Research Institute, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chengjun Sui
- Department of Special Treatment, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Cheng Yang
- Department of Special Treatment, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
9
|
Khokhar M, Purohit P. The emerging role of T helper 9 (Th9) cells in immunopathophysiology: A comprehensive review of their effects and responsiveness in various disease states. Int Rev Immunol 2024; 43:341-360. [PMID: 38864109 DOI: 10.1080/08830185.2024.2364586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/10/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024]
Abstract
Th9 cells, a subset of T-helper cells producing interleukin-9 (IL-9), play a vital role in the adaptive immune response and have diverse effects in different diseases. Regulated by transcription factors like PU.1 and IRF4, and cytokines such as IL-4 and TGF-β, Th9 cells drive tissue inflammation. This review focuses on their emerging role in immunopathophysiology. Th9 cells exhibit immune-mediated cancer cell destruction, showing promise in glioma and cervical cancer treatment. However, their role in breast and lung cancer is intricate, requiring a deeper understanding of pro- and anti-tumor aspects. Th9 cells, along with IL-9, foster T cell and immune cell proliferation, contributing to autoimmune disorders. They are implicated in psoriasis, atopic dermatitis, and infections. In allergic reactions and asthma, Th9 cells fuel pro-inflammatory responses. Targeting Foxo1 may regulate innate and adaptive immune responses, alleviating disease symptoms. This comprehensive review outlines Th9 cells' evolving immunopathophysiological role, emphasizing the necessity for further research to grasp their effects and potential therapeutic applications across diseases.
Collapse
Affiliation(s)
- Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Purvi Purohit
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| |
Collapse
|
10
|
Gonzalez-Avalos E, Onodera A, Samaniego-Castruita D, Rao A, Ay F. Predicting gene expression state and prioritizing putative enhancers using 5hmC signal. Genome Biol 2024; 25:142. [PMID: 38825692 PMCID: PMC11145787 DOI: 10.1186/s13059-024-03273-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/11/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Like its parent base 5-methylcytosine (5mC), 5-hydroxymethylcytosine (5hmC) is a direct epigenetic modification of cytosines in the context of CpG dinucleotides. 5hmC is the most abundant oxidized form of 5mC, generated through the action of TET dioxygenases at gene bodies of actively-transcribed genes and at active or lineage-specific enhancers. Although such enrichments are reported for 5hmC, to date, predictive models of gene expression state or putative regulatory regions for genes using 5hmC have not been developed. RESULTS Here, by using only 5hmC enrichment in genic regions and their vicinity, we develop neural network models that predict gene expression state across 49 cell types. We show that our deep neural network models distinguish high vs low expression state utilizing only 5hmC levels and these predictive models generalize to unseen cell types. Further, in order to leverage 5hmC signal in distal enhancers for expression prediction, we employ an Activity-by-Contact model and also develop a graph convolutional neural network model with both utilizing Hi-C data and 5hmC enrichment to prioritize enhancer-promoter links. These approaches identify known and novel putative enhancers for key genes in multiple immune cell subsets. CONCLUSIONS Our work highlights the importance of 5hmC in gene regulation through proximal and distal mechanisms and provides a framework to link it to genome function. With the recent advances in 6-letter DNA sequencing by short and long-read techniques, profiling of 5mC and 5hmC may be done routinely in the near future, hence, providing a broad range of applications for the methods developed here.
Collapse
Affiliation(s)
- Edahi Gonzalez-Avalos
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
- Bioinformatics and Systems Biology Graduate Program, University of California San Diego, La Jolla, CA, 92093, USA
| | - Atsushi Onodera
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Daniela Samaniego-Castruita
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, 92093, USA
| | - Anjana Rao
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA.
- Bioinformatics and Systems Biology Graduate Program, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA.
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Ferhat Ay
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA.
- Bioinformatics and Systems Biology Graduate Program, University of California San Diego, La Jolla, CA, 92093, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
11
|
Son A, Baral I, Falduto GH, Schwartz DM. Locus of (IL-9) control: IL9 epigenetic regulation in cellular function and human disease. Exp Mol Med 2024; 56:1331-1339. [PMID: 38825637 PMCID: PMC11263352 DOI: 10.1038/s12276-024-01241-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 06/04/2024] Open
Abstract
Interleukin-9 (IL-9) is a multifunctional cytokine with roles in a broad cross-section of human diseases. Like many cytokines, IL-9 is transcriptionally regulated by a group of noncoding regulatory elements (REs) surrounding the IL9 gene. These REs modulate IL-9 transcription by forming 3D loops that recruit transcriptional machinery. IL-9-promoting transcription factors (TFs) can bind REs to increase locus accessibility and permit chromatin looping, or they can be recruited to already accessible chromatin to promote transcription. Ample mechanistic and genome-wide association studies implicate this interplay between IL-9-modulating TFs and IL9 cis-REs in human physiology, homeostasis, and disease.
Collapse
Affiliation(s)
- Aran Son
- Neuroscience Department, International School for Advanced Studies (SISSA), via Bonomea 265, Trieste, 34136, Italy
| | - Ishita Baral
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guido H Falduto
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniella M Schwartz
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Tyagi S, Sadhu S, Sharma T, Paul A, Pandey M, Nain VK, Rathore DK, Chatterjee S, Awasthi A, Pandey AK. VapC12 ribonuclease toxin modulates host immune response during Mycobacterium tuberculosis infection. Front Immunol 2024; 15:1302163. [PMID: 38515752 PMCID: PMC10955575 DOI: 10.3389/fimmu.2024.1302163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 02/01/2024] [Indexed: 03/23/2024] Open
Abstract
Mechanistic understanding of antibiotic persistence is a prerequisite in controlling the emergence of MDR cases in Tuberculosis (TB). We have reported that the cholesterol-induced activation of VapC12 ribonuclease is critical for disease persistence in TB. In this study, we observed that relative to the wild type, mice infected with ΔvapC12 induced a pro-inflammatory response, had a higher pathogen load, and responded better to the anti-TB treatment. In a high-dose infection model, all the mice infected with ΔvapC12 succumbed early to the disease. Finally, we reported that the above phenotype of ΔvapC12 was dependent on the presence of the TLR4 receptor. Overall, the data suggests that failure of a timely resolution of the early inflammation by the ΔvapC12 infected mice led to hyperinflammation, altered T-cell response and high bacterial load. In conclusion, our findings suggest the role of the VapC12 toxin in modulating the innate immune response of the host in ways that favor the long-term survival of the pathogen inside the host.
Collapse
Affiliation(s)
- Shaifali Tyagi
- Mycobacterial Pathogenesis Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Srikanth Sadhu
- Immunobiology Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Taruna Sharma
- Mycobacterial Pathogenesis Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Abhijit Paul
- Complex Analysis Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Manitosh Pandey
- Mycobacterial Pathogenesis Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Vaibhav Kumar Nain
- Mycobacterial Pathogenesis Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Deepak Kumar Rathore
- Immunobiology Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Samrat Chatterjee
- Complex Analysis Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Amit Awasthi
- Immunobiology Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Amit Kumar Pandey
- Mycobacterial Pathogenesis Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
13
|
Roostaee A, Yaghobi R, Afshari A, Jafarinia M. Regulatory role of T helper 9/interleukin-9: Transplantation view. Heliyon 2024; 10:e26359. [PMID: 38420400 PMCID: PMC10900956 DOI: 10.1016/j.heliyon.2024.e26359] [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: 07/19/2023] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
T helper 9 (Th9) cells, a subset of CD4+ T helper cells, have emerged as a valuable target for immune cell therapy due to their potential to induce immunomodulation and tolerance. The Th9 cells mainly produce interleukin (IL)-9 and are known for their defensive effects against helminth infections, allergic and autoimmune responses, and tumor suppression. This paper explores the mechanisms involved in the generation and differentiation of Th9 cells, including the cytokines responsible for their polarization and stabilization, the transcription factors necessary for their differentiation, as well as the role of Th9 cells in inflammatory and autoimmune diseases, allergic reactions, and cancer immunotherapies. Recent research has shown that the differentiation of Th9 cells is coregulated by the transcription factors transforming growth factor β (TGF-β), IL-4, and PU.1, which are also known to secrete IL-10 and IL-21. Multiple cell types, such as T and B cells, mast cells, and airway epithelial cells, are influenced by IL-9 due to its pleiotropic effects.
Collapse
Affiliation(s)
- Azadeh Roostaee
- Department of Genetics, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Ramin Yaghobi
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afsoon Afshari
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojtaba Jafarinia
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| |
Collapse
|
14
|
Sadhu S, Dandotiya J, Dalal R, Khatri R, Mykytyn AZ, Batra A, Kaur M, Chandwaskar R, Singh V, Kamboj A, Srivastava M, Mani S, Asthana S, Samal S, Rizvi ZA, Salunke DB, Haagmans BL, Awasthi A. Fangchinoline inhibits SARS-CoV-2 and MERS-CoV entry. Antiviral Res 2023; 220:105743. [PMID: 37949319 DOI: 10.1016/j.antiviral.2023.105743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2, lead to mild to severe respiratory illness and resulted in 6.9 million deaths worldwide. Although vaccines are effective in preventing COVID-19, they may not be sufficient to protect immunocompromised individuals from this respiratory illness. Moreover, novel emerging variants of SARS-CoV-2 pose a risk of new COVID-19 waves. Therefore, identification of effective antivirals is critical in controlling SARS and other coronaviruses, such as MERS-CoV. We show that Fangchinoline (Fcn), a bisbenzylisoquinoline alkaloid, inhibits replication of SARS-CoV, SARS-CoV-2, and MERS-CoV in a range of in vitro assays, by blocking entry. Therapeutic use of Fcn inhibited viral loads in the lungs, and suppressed associated airway inflammation in hACE2. Tg mice and Syrian hamster infected with SARS-CoV-2. Combination of Fcn with remdesivir (RDV) or an anti-leprosy drug, Clofazimine, exhibited synergistic antiviral activity. Compared to Fcn, its synthetic derivative, MK-04-003, more effectively inhibited SARS-CoV-2 and its variants B.1.617.2 and BA.5 in mice. Taken together these data demonstrate that Fcn is a pan beta coronavirus inhibitor, which possibly can be used to combat novel emerging coronavirus diseases.
Collapse
Affiliation(s)
- Srikanth Sadhu
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India; Immunology-Core Lab, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Jyotsna Dandotiya
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Rajdeep Dalal
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Ritika Khatri
- Infection and Immunology Center, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Anna Z Mykytyn
- Viroscience Department, Erasmus University Medical Center, Netherlands; Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Netherlands
| | - Aashima Batra
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Manpreet Kaur
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | | | - Virendra Singh
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Aarzoo Kamboj
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Mitul Srivastava
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Shailendra Mani
- Infection and Immunology Center, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Shailendra Asthana
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Sweety Samal
- Infection and Immunology Center, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Zaigham Abbas Rizvi
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India; Immunology-Core Lab, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Deepak B Salunke
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Bart L Haagmans
- Viroscience Department, Erasmus University Medical Center, Netherlands
| | - Amit Awasthi
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India; Immunology-Core Lab, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| |
Collapse
|
15
|
Lee WH, Hong KJ, Li H, Lee GR. Transcription Factor Id1 Plays an Essential Role in Th9 Cell Differentiation by Inhibiting Tcf3 and Tcf4. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2305527. [PMID: 37867222 PMCID: PMC10724384 DOI: 10.1002/advs.202305527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/08/2023] [Indexed: 10/24/2023]
Abstract
T helper type 9 (Th9) cells play important roles in immune responses by producing interleukin-9 (IL-9). Several transcription factors are responsible for Th9 cell differentiation; however, transcriptional regulation of Th9 cells is not fully understood. Here, it is shown that Id1 is an essential transcriptional regulator of Th9 cell differentiation. Id1 is induced by IL-4 and TGF-β. Id1-deficient naïve CD4 T cells fail to differentiate into Th9 cells, and overexpression of Id1 induce expression of IL-9. Mass spectrometry analysis reveals that Id1 interacts with Tcf3 and Tcf4 in Th9 cells. In addition, RNA-sequencing, chromatin immunoprecipitation, and transient reporter assay reveal that Tcf3 and Tcf4 bind to the promoter region of the Il9 gene to suppress its expression, and that Id1 inhibits their function, leading to Th9 differentiation. Finally, Id1-deficient Th9 cells ameliorate airway inflammation in an animal model of asthma. Thus, Id1 is a transcription factor that plays an essential role in Th9 cell differentiation by inhibiting Tcf3 and Tcf4.
Collapse
Affiliation(s)
- Woo Ho Lee
- Department of Life ScienceSogang University35 Baekbeom‐roMapo‐guSeoul04107South Korea
| | - Kyung Jin Hong
- Department of Life ScienceSogang University35 Baekbeom‐roMapo‐guSeoul04107South Korea
| | - Hua‐Bing Li
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of Medicine280 Chongqing South Rd, Building #5‐602Shanghai200025China
| | - Gap Ryol Lee
- Department of Life ScienceSogang University35 Baekbeom‐roMapo‐guSeoul04107South Korea
| |
Collapse
|
16
|
Bian X, Xue H, Jing D, Wang Y, Zhou G, Zhu F. Role of Serum/Glucocorticoid-Regulated Kinase 1 (SGK1) in Immune and Inflammatory Diseases. Inflammation 2023; 46:1612-1625. [PMID: 37353719 DOI: 10.1007/s10753-023-01857-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023]
Abstract
Serum/glucocorticoid-regulated kinase 1 (SGK1), a member of the serine/threonine protein kinase gene family, is primarily regulated by serum and glucocorticoids. SGK1 is involved in the development of tumors and fibrotic diseases. However, relatively little research has been conducted on their role in immune and inflammatory diseases. SGK1 may act as a pivotal immune regulatory gene by modulating immune cells (e.g., T cells, macrophages, dendritic cells, and neutrophils) and functions and is involved in the pathogenesis of some immune and inflammatory diseases, such as inflammatory bowel disease, multiple sclerosis, allergic diseases, sepsis, and major depressive disorder. This review aims to provide an overview of the latest research focusing on the immune and inflammatory regulatory roles of SGK1 and provide new insights into diagnostic and therapeutic approaches for immune and inflammatory diseases.
Collapse
Affiliation(s)
- Xixi Bian
- Clinical Medical College of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Honglu Xue
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Dehuai Jing
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Yan Wang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Guangxi Zhou
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China.
| | - Fengqin Zhu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China.
| |
Collapse
|
17
|
Chen J, Zhang Y, Zhang H, Zhang M, Dong H, Qin T, Gao S, Wang S. IL-24 is the key effector of Th9 cell-mediated tumor immunotherapy. iScience 2023; 26:107531. [PMID: 37680459 PMCID: PMC10480301 DOI: 10.1016/j.isci.2023.107531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/28/2023] [Accepted: 07/28/2023] [Indexed: 09/09/2023] Open
Abstract
Th9 cells are powerful effector T cells for cancer immunotherapy. However, the underlying antitumor mechanism of Th9 cells still needs to be further elucidated. Here, we show that Th9 cells express high levels of not only IL-9, but also IL-24. We found that knockout of Il24 gene in Th9 cells promotes Th9 cell proliferation in vitro, but decreases Th9 cell survival in vitro and in vivo. Interestingly, knockout of Il24 gene in Th9 cells decreases the tumor-specific cytotoxicity of Th9 cells in vitro. In addition, immunotherapy with Il24 knockout Th9 cells exhibit less tumor inhibition than regular Th9 cells in mouse tumor models. We found that inhibition of Foxo1 by a specific inhibitor downregulates IL-24 expression in Th9 cells and decreases Th9 cell antitumor efficacy in vivo. Our results identify IL-24 as a powerful antitumor effector of Th9 cells and provide a target in Th9 cell-mediated tumor therapy.
Collapse
Affiliation(s)
- Jintong Chen
- Department of Cancer Immunology, First Hospital of Jilin University, Changchun 130061, China
| | - Yunwei Zhang
- Department of Hematology, First Hospital of Jilin University, Changchun 130061, China
| | - Hua Zhang
- Department of Gastrointestinal Surgery, First Hospital of Jilin University, Changchun 130021, China
| | - Mingyue Zhang
- Department of Gynecological Oncology, First Hospital of Jilin University, Changchun 130021, China
| | - He Dong
- Department of Gynecological Oncology, First Hospital of Jilin University, Changchun 130021, China
| | - Tianxue Qin
- Department of Hematology, First Hospital of Jilin University, Changchun 130061, China
| | - Sujun Gao
- Department of Hematology, First Hospital of Jilin University, Changchun 130061, China
| | - Siqing Wang
- Department of Cancer Immunology, First Hospital of Jilin University, Changchun 130061, China
| |
Collapse
|
18
|
Rizvi ZA, Dandotiya J, Sadhu S, Khatri R, Singh J, Singh V, Adhikari N, Sharma K, Das V, Pandey AK, Das B, Medigeshi G, Mani S, Bhatnagar S, Samal S, Pandey AK, Garg PK, Awasthi A. Omicron sub-lineage BA.5 infection results in attenuated pathology in hACE2 transgenic mice. Commun Biol 2023; 6:935. [PMID: 37704701 PMCID: PMC10499788 DOI: 10.1038/s42003-023-05263-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 08/20/2023] [Indexed: 09/15/2023] Open
Abstract
A recently emerged sub-lineage of Omicron, BA.5, together with BA.4, caused a fifth wave of coronavirus disease (COVID-19) in South Africa and subsequently emerged as a predominant strain globally due to its high transmissibility. The lethality of BA.5 infection has not been studied in an acute hACE2 transgenic (hACE2.Tg) mouse model. Here, we investigated tissue-tropism and immuno-pathology induced by BA.5 infection in hACE2.Tg mice. Our data show that intranasal infection of BA.5 in hACE2.Tg mice resulted in attenuated pulmonary infection and pathology with diminished COVID-19-induced clinical and pathological manifestations. BA.5, similar to Omicron (B.1.1.529), infection led to attenuated production of inflammatory cytokines, anti-viral response and effector T cell response as compared to the ancestral strain of SARS-CoV-2, Wuhan-Hu-1. We show that mice recovered from B.1.1.529 infection showed robust protection against BA.5 infection associated with reduced lung viral load and pathology. Together, our data provide insights as to why BA.5 infection escapes previous SARS-CoV-2 exposure induced-T cell immunity but may result in milder immuno-pathology and alleviated chances of re-infectivity in Omicron-recovered individuals.
Collapse
Affiliation(s)
- Zaigham Abbas Rizvi
- Centre for Immuno-biology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India.
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India.
| | - Jyotsna Dandotiya
- Centre for Immuno-biology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Srikanth Sadhu
- Centre for Immuno-biology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Ritika Khatri
- Centre for Viral Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Janmejay Singh
- Bioassay Laboratory, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001, India
| | - Virendra Singh
- Centre for Immuno-biology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Neeta Adhikari
- Centre for Immuno-biology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Kritika Sharma
- Centre for Immuno-biology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Vinayake Das
- Centre for Immuno-biology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Amit Kumar Pandey
- Centre for Tuberculosis and Bacterial Diseases Research, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Bhabatosh Das
- Centre for Microbiome and Anti-Microbial Resistance, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Guruprasad Medigeshi
- Bioassay Laboratory, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001, India
| | - Shalendra Mani
- Centre for Viral Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Shinjini Bhatnagar
- Centre for Maternal and Child Health, Translational Health Science and Technology NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Sweety Samal
- Centre for Viral Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India
| | - Anil Kumar Pandey
- Department of Physiology, ESIC Medical College & Hospital, Faridabad, 121001, India
| | - Pramod Kumar Garg
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Amit Awasthi
- Centre for Immuno-biology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India.
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, 121001, India.
| |
Collapse
|
19
|
Sadhu S, Dalal R, Dandotiya J, Binayke A, Singh V, Tripathy MR, Das V, Goswami S, Kumar S, Rizvi ZA, Awasthi A. IL-9 aggravates SARS-CoV-2 infection and exacerbates associated airway inflammation. Nat Commun 2023; 14:4060. [PMID: 37429848 DOI: 10.1038/s41467-023-39815-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 06/30/2023] [Indexed: 07/12/2023] Open
Abstract
SARS-CoV-2 infection is known for causing broncho-alveolar inflammation. Interleukin 9 (IL-9) induces airway inflammation and bronchial hyper responsiveness in respiratory viral illnesses and allergic inflammation, however, IL-9 has not been assigned a pathologic role in COVID-19. Here we show, in a K18-hACE2 transgenic (ACE2.Tg) mouse model, that IL-9 contributes to and exacerbates viral spread and airway inflammation caused by SARS-CoV-2 infection. ACE2.Tg mice with CD4+ T cell-specific deficiency of the transcription factor Forkhead Box Protein O1 (Foxo1) produce significantly less IL-9 upon SARS-CoV-2 infection than the wild type controls and they are resistant to the severe inflammatory disease that characterises the control mice. Exogenous IL-9 increases airway inflammation in Foxo1-deficient mice, while IL-9 blockade reduces and suppresses airway inflammation in SARS-CoV-2 infection, providing further evidence for a Foxo1-Il-9 mediated Th cell-specific pathway playing a role in COVID-19. Collectively, our study provides mechanistic insight into an important inflammatory pathway in SARS-CoV-2 infection, and thus represents proof of principle for the development of host-directed therapeutics to mitigate disease severity.
Collapse
Affiliation(s)
- Srikanth Sadhu
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
- Immunology-Core Laboratory, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
| | - Rajdeep Dalal
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
| | - Jyotsna Dandotiya
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
| | - Akshay Binayke
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
| | - Virendra Singh
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
| | - Manas Ranjan Tripathy
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
- Immunology-Core Laboratory, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
| | - Vinayaka Das
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
| | - Sandeep Goswami
- Immunology-Core Laboratory, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
| | - Shakti Kumar
- Centre for Human Microbiome and Anti-Microbial Resistance, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001, Haryana, India
| | - Zaigham Abbas Rizvi
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
- Immunology-Core Laboratory, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India
| | - Amit Awasthi
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India.
- Immunology-Core Laboratory, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121 001, Haryana, India.
| |
Collapse
|
20
|
Ren H, Chen Y, Zhu Z, Xia J, Liu S, Hu Y, Qin X, Zhang L, Ding Y, Xia S, Wang J. FOXO1 regulates Th17 cell-mediated hepatocellular carcinoma recurrence after hepatic ischemia-reperfusion injury. Cell Death Dis 2023; 14:367. [PMID: 37330523 PMCID: PMC10276824 DOI: 10.1038/s41419-023-05879-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/07/2023] [Accepted: 05/31/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Hepatic ischemia-reperfusion injury (IRI) is considered as an effecting factor for hepatocellular carcinoma (HCC) recurrence. Th17/Treg cells are a pair of essential components in adaptive immune response in liver IRI, and forkhead box O1 (FOXO1) has the properties of maintaining the function and phenotype of immune cells. Herein, we illuminated the correlation and function between Th17/Treg cell balance and FOXO1 in IRI-induced HCC recurrence. METHODS RNA sequencing was performed on naive CD4+ T cells from normal and IRI model mice to identify relevant transcription factors. Western blotting, qRT-PCR, immunohistochemical staining, and flow cytometry were performed in IRI models to indicate the effect of FOXO1 on the polarization of Th17/Treg cells. Then, transwell assay of HCC cell migration and invasion, clone formation, wound healing assay, and Th17 cells adoptively transfer was utilized to assess the function of Th17 cells in IRI-induced HCC recurrence in vitro and in vivo. RESULTS Owning to the application of RNA sequencing, FOXO1 was screened and assumed to perform a significant function in hepatic IRI. The IRI model demonstrated that up-regulation of FOXO1 alleviated IR stress by attenuating inflammatory stress, maintaining microenvironment homeostasis, and reducing the polarization of Th17 cells. Mechanistically, Th17 cells accelerated IRI-induced HCC recurrence by shaping the hepatic pre-metastasis microenvironment, activating the EMT program, promoting cancer stemness and angiogenesis, while the upregulation of FOXO1 can stabilize the liver microenvironment homeostasis and alleviate the negative effects of Th17 cells. Moreover, the adoptive transfer of Th17 cells in vivo revealed its inducing function in IRI-induced HCC recurrence. CONCLUSIONS These results indicated that FOXO1-Th17/Treg axis exerts a crucial role in IRI-mediated immunologic derangement and HCC recurrence, which could be a promising target for reducing the HCC recurrence after hepatectomy. Liver IRI affects the balance of Th17/Treg cells by inhibiting the expression of FOXO1, and the increase of Th17 cells has the ability to induce HCC recurrence through EMT program, cancer stemness pathway, the formation of premetastatic microenvironment and angiogenesis.
Collapse
Grants
- the National Natural Science Foundation of China (82270646),the Fundamental Research Funds for the Central Universities (0214-14380510), the Nanjing health science and technology development project for Distinguished Young Scholars,the Nanjing health science and technology development project for Distinguished Young Scholars (JQX19002),Project of Modern Hospital Management and Development Institute, Nanjing University and Aid project of Nanjing Drum Tower Hospital Health, Education & Research Foundation(NDYG2022057),fundings for Clinical Trials from the Affiliated Drum Tower Hospital, Medical School of Nanjing University(2022-LCYJ-PY-35),the Chen Xiao-ping Foundation for the Development of Science and Technology of Hubei Province, China (CXPJJH121001-2021073)
Collapse
Affiliation(s)
- Haozhen Ren
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu Province, China
| | - Yuyan Chen
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Zhengyi Zhu
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Jinkun Xia
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Shujun Liu
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yingzhe Hu
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Xueqian Qin
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Lu Zhang
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yitao Ding
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu Province, China
| | - Senzhe Xia
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu Province, China.
| | - Jinglin Wang
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
21
|
Wang YH, Peng YJ, Liu FC, Lin GJ, Huang SH, Sytwu HK, Cheng CP. Interleukin 26 Induces Macrophage IL-9 Expression in Rheumatoid Arthritis. Int J Mol Sci 2023; 24:ijms24087526. [PMID: 37108686 PMCID: PMC10139149 DOI: 10.3390/ijms24087526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/15/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with chronic inflammation, bone erosion, and joint deformation. Synovial tissue in RA patients is full of proinflammatory cytokines and infiltrated immune cells, such as T help (Th) 9, Th17, macrophages, and osteoclasts. Recent reports emphasized a new member of the interleukin (IL)-10 family, IL-26, an inducer of IL-17A that is overexpressed in RA patients. Our previous works found that IL-26 inhibits osteoclastogenesis and conducts monocyte differentiation toward M1 macrophages. In this study, we aimed to clarify the effect of IL-26 on macrophages linking to Th9 and Th17 in IL-9 and IL-17 expression and downstream signal transduction. Murine and human macrophage cell lines and primary culture cells were used and stimulated by IL26. Cytokines expressions were evaluated by flow cytometry. Signal transduction and transcription factors expression were detected by Western blot and real time-PCR. Our results show that IL-26 and IL-9 colocalized in macrophage in RA synovium. IL-26 directly induces macrophage inflammatory cytokines IL-9 and IL-17A expression. IL-26 increases the IL-9 and IL-17A upstream mechanisms IRF4 and RelB expression. Moreover, the AKT-FoxO1 pathway is also activated by IL-26 in IL-9 and IL-17A expressing macrophage. Blockage of AKT phosphorylation enhances IL-26 stimulating IL-9-producing macrophage cells. In conclusion, our results support that IL-26 promotes IL-9- and IL-17-expressing macrophage and might initiate IL-9- and IL-17-related adaptive immunity in rheumatoid arthritis. Targeting IL-26 may a potential therapeutic strategy for rheumatoid arthritis or other IL-9 plus IL-17 dominant diseases.
Collapse
Affiliation(s)
- Yi-Hsun Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan
| | - Yi-Jen Peng
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
| | - Feng-Cheng Liu
- Division of Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
| | - Gu-Jiun Lin
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei 11490, Taiwan
| | - Shing-Hwa Huang
- Division of Breast Surgery, Department of Surgery, New Taipei City Hospital, New Taipei City 241204, Taiwan
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Chia-Pi Cheng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei 11490, Taiwan
| |
Collapse
|
22
|
Cui X, Zhao H, Wei S, Du Q, Dong K, Yan Y, Geller DA. Hepatocellular carcinoma-derived FOXO1 inhibits tumor progression by suppressing IL-6 secretion from macrophages. Neoplasia 2023; 40:100900. [PMID: 37058885 PMCID: PMC10123375 DOI: 10.1016/j.neo.2023.100900] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/16/2023]
Abstract
Tumor heterogeneity dominates tumor biological behavior and shapes the tumor microenvironment. However, the mechanisms of tumor genetic features modulate immunity response were not clearly clarified. Tumor associated macrophages (TAMs) exert distinct immune functions in the progression of hepatocellular carcinoma (HCC) based on the inducible phenotype. FOXO family members sense changes in the extracellular or intracellular environment by activating a series of signaling pathways. FOXO1, a transcription factor that a common suppressor in hepatocellular carcinoma, correlated with a better tumor biological behavior in HCC through shaping macrophages anti-tumour response. Here, we found that human HCC tissue microarray (TMA) slides were employed to showed tumor derived FOXO1 negatively related with distribution of protumour macrophages. This phenomenon was confirmed in mouse xenograft model and in vitro. HCC-derived FOXO1 inhibits tumorigenesis not only by targeting tumor cells but also by synchronizing with re-educated macrophages. These effects may be partially dependent on FOXO1 transcriptionally modulates IRF-1/nitrio oxide (NO) axis in exerting effects in macrophages and decreasing IL-6 releasing from macrophages in tumor microenvironment indirectly. This feedback suppressed the progression of HCC by inactivation of IL-6/STAT3 in HCC. It implicates the potential role of FOXO1 in the therapeutic effects for modulating immune response by targeting macrophages.
Collapse
Affiliation(s)
- Xiao Cui
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China; Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15260, USA.
| | - Huiyong Zhao
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
| | - Sheng Wei
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
| | - Qiang Du
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15260, USA.
| | - Kun Dong
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Yihe Yan
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China.
| | - David A Geller
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
23
|
Crosstalk of Transcriptional Regulators of Adaptive Immune System and microRNAs: An Insight into Differentiation and Development. Cells 2023; 12:cells12040635. [PMID: 36831302 PMCID: PMC9953855 DOI: 10.3390/cells12040635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/27/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
MicroRNAs (miRNAs), as small regulatory RNA molecules, are involved in gene expression at the post-transcriptional level. Hence, miRNAs contribute to gene regulation of various steps of different cell subsets' differentiation, maturation, and activation. The adaptive immune system arm, which exhibits the most specific immune responses, is also modulated by miRNAs. The generation and maturation of various T-cell subsets concomitant with B-cells is under precise regulation of miRNAs which function directly on the hallmark genes of each cell subset or indirectly through regulation of signaling pathway mediators and/or transcription factors involved in this maturation journey. In this review, we first discussed the origination process of common lymphocyte progenitors from hematopoietic stem cells, which further differentiate into various T-cell subsets under strict regulation of miRNAs and transcription factors. Subsequently, the differentiation of B-cells from common lymphocyte progenitors in bone marrow and periphery were discussed in association with a network of miRNAs and transcription factors.
Collapse
|
24
|
Li Y, Li L, Zhang W, Gao Y. Amphiregulin/epidermal growth factor receptor/hypoxia-inducible factor-1α pathway regulates T helper 9 and T cytotoxic 9 cell response in adult patients with infectious mononucleosis. BIOMOLECULES AND BIOMEDICINE 2023; 23:63-72. [PMID: 36154925 PMCID: PMC9901907 DOI: 10.17305/bjbms.2022.8013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/16/2022] [Indexed: 02/03/2023]
Abstract
Amphiregulin (AREG)/epidermal growth factor receptor (EGFR) signaling induces hypoxia-inducible factor-1α (HIF-1α), leading to promotion of T helper 9 (Th9) differentiation and anti-tumor functions. However, the role of the AREG/EGFR/HIF-1α pathway in regulating interleukin-9 (IL-9) production by T cells in adult patients with infectious mononucleosis (IM) has not been fully elucidated. Fifty IM patients and 20 controls were enrolled. The percentages of Th9 and T cytotoxic 9 (Tc9) cells, the mRNA relative expressions of the transcription factors of IL-9-secreting T cells, purine-rich nucleic acid binding protein 1 (PU.1) and forkhead box protein O1 (FOXO1), and the levels of IL-9, AREG, EGFR, and HIF-1α were measured. Peripheral blood mononuclear cells from IM patients were stimulated with EGFR inhibitor or exogenous AREG in the presence or absence of anti-HIF-1α. Regulation of the AREG/EGFR/HIF-1α pathway to IL-9 production by T cells was assessed. The percentages of Th9 and Tc9 cells, plasma IL-9 levels, and PU.1 and FOXO1 mRNA expressions were elevated in IM patients. Plasma levels of AREG and HIF-1α were also increased in IM patients. AREG levels correlated positively with the percentages of Th9 and Tc9 cells in IM patients. Inhibition of EGFR suppressed IL-9-producing T cell differentiation and HIF-1α production. Exogenous AREG stimulation not only induced EGFR and HIF-1α expression but also promoted IL-9-secreting T cell differentiation. Neutralization of HIF-1α abrogated AREG/EGFR-induced Th9 and Tc9 differentiation in IM patients. The current data suggested that the AREG/EGFR/HIF-1α pathway contributed to the elevation of Th9 and Tc9 differentiation in IM patients.
Collapse
Affiliation(s)
- Yu Li
- Department of Infectious Diseases, Shaanxi Provincial People’s Hospital, The Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi Province, China
| | - Lan Li
- Department of Hematology, Shaanxi Provincial People’s Hospital, The Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi Province, China
| | - Weihua Zhang
- Department of Hematology, Shaanxi Provincial People’s Hospital, The Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi Province, China
| | - Ying Gao
- Department of Hematology, Shaanxi Provincial People’s Hospital, The Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi Province, China,Correspondence to Ying Gao:
| |
Collapse
|
25
|
Rizvi ZA, Babele P, Madan U, Sadhu S, Tripathy MR, Goswami S, Mani S, Dikshit M, Awasthi A. Pharmacological potential of Withania somnifera (L.) Dunal and Tinospora cordifolia (Willd.) Miers on the experimental models of COVID-19, T cell differentiation, and neutrophil functions. Front Immunol 2023; 14:1138215. [PMID: 36960064 PMCID: PMC10028191 DOI: 10.3389/fimmu.2023.1138215] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/20/2023] [Indexed: 03/09/2023] Open
Abstract
Cytokine release syndrome (CRS) due to severe acute respiratory coronavirus-2 (SARS-CoV-2) infection leads to life-threatening pneumonia which has been associated with coronavirus disease (COVID-19) pathologies. Centuries-old Asian traditional medicines such as Withania somnifera (L.) Dunal (WS) and Tinospora cordifolia (Willd.) Miers (TC) possess potent immunomodulatory effects and were used by the AYUSH ministry, in India during the COVID-19 pandemic. In the present study, we investigated WS and TC's anti-viral and immunomodulatory efficacy at the human equivalent doses using suitable in vitro and in vivo models. While both WS and TC showed immuno-modulatory potential, WS showed robust protection against loss in body weight, viral load, and pulmonary pathology in the hamster model of SARS-CoV2. In vitro pretreatment of mice and human neutrophils with WS and TC had no adverse effect on PMA, calcium ionophore, and TRLM-induced ROS generation, phagocytosis, bactericidal activity, and NETs formation. Interestingly, WS significantly suppressed the pro-inflammatory cytokines-induced Th1, Th2, and Th17 differentiation. We also used hACE2 transgenic mice to further investigate the efficacy of WS against acute SARS-CoV2 infection. Prophylactic treatment of WS in the hACE2 mice model showed significant protection against body weight loss, inflammation, and the lung viral load. The results obtained indicate that WS promoted the immunosuppressive environment in the hamster and hACE2 transgenic mice models and limited the worsening of the disease by reducing inflammation, suggesting that WS might be useful against other acute viral infections. The present study thus provides pre-clinical efficacy data to demonstrate a robust protective effect of WS against COVID-19 through its broader immunomodulatory activity.
Collapse
Affiliation(s)
- Zaigham Abbas Rizvi
- Immuno-biology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- *Correspondence: Amit Awasthi, ; Madhu Dikshit, ; ; Zaigham Abbas Rizvi,
| | - Prabhakar Babele
- NCD, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Upasna Madan
- Immuno-biology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Srikanth Sadhu
- Immuno-biology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Manas Ranjan Tripathy
- Immuno-biology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Sandeep Goswami
- Immuno-biology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Shailendra Mani
- NCD, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Madhu Dikshit
- NCD, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, Faridabad, Haryana, India
- Pharmacology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- *Correspondence: Amit Awasthi, ; Madhu Dikshit, ; ; Zaigham Abbas Rizvi,
| | - Amit Awasthi
- Immuno-biology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- *Correspondence: Amit Awasthi, ; Madhu Dikshit, ; ; Zaigham Abbas Rizvi,
| |
Collapse
|
26
|
CD4+IL9+ (Th9) cells as the major source of IL-9, potentially modulate Th17/Treg mediated host immune response during experimental cerebral malaria. Mol Immunol 2022; 152:240-254. [DOI: 10.1016/j.molimm.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/18/2022] [Accepted: 11/07/2022] [Indexed: 11/15/2022]
|
27
|
Kitamoto T, Lee YK, Sultana N, Watanabe H, McKimpson WM, Du W, Fan J, Diaz B, Lin HV, Leibel RL, Belvedere S, Accili D, Accili D. Chemical induction of gut β-like-cells by combined FoxO1/Notch inhibition as a glucose-lowering treatment for diabetes. Mol Metab 2022; 66:101624. [PMID: 36341906 PMCID: PMC9664469 DOI: 10.1016/j.molmet.2022.101624] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE Lifelong insulin replacement remains the mainstay of type 1 diabetes treatment. Genetic FoxO1 ablation promotes enteroendocrine cell (EECs) conversion into glucose-responsive β-like cells. Here, we tested whether chemical FoxO1 inhibitors can generate β-like gut cells. METHODS We used Ngn3-or Villin-driven FoxO1 ablation to capture the distinctive developmental effects of FoxO1 on EEC pool. We combined FoxO1 ablation with Notch inhibition to enhance the expansion of EEC pool. We tested the ability of an orally available small molecule of FoxO1 inhibitor, Cpd10, to phenocopy genetic ablation of FoxO1. We evaluated the therapeutic impact of genetic ablation or chemical inhibition of FoxO1 on insulin-deficient diabetes in Ins2Akita/+ mice. RESULTS Pan-intestinal epithelial FoxO1 ablation expanded the EEC pool, induced β-like cells, and improved glucose tolerance in Ins2Akita/+ mice. This genetic effect was phenocopied by Cpd10. Cpd10 induced β-like cells that released insulin in response to glucose in gut organoids, and this effect was enhanced by the Notch inhibitor, DBZ. In Ins2Akita/+ mice, a five-day course of either Cpd10 or DBZ induced intestinal insulin-immunoreactive β-like cells, lowered glycemia, and increased plasma insulin levels without apparent adverse effects. CONCLUSION These results provide proof of principle of gut cell conversion into β-like cells by a small molecule FoxO1 inhibitor, paving the way for clinical applications.
Collapse
Affiliation(s)
- Takumi Kitamoto
- Department of Medicine and Columbia University, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA; Chiba University Graduate School of Medicine, Chiba, Japan, 2608670.
| | | | - Nishat Sultana
- Department of Pediatrics Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Hitoshi Watanabe
- Department of Medicine and Columbia University, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
| | - Wendy M McKimpson
- Department of Medicine and Columbia University, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
| | - Wen Du
- Department of Medicine and Columbia University, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
| | - Jason Fan
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Miami, FL, 33146, USA
| | - Bryan Diaz
- Department of Pediatrics Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Hua V Lin
- BioFront Therapeutics, Beijing, China
| | - Rudolph L Leibel
- Department of Pediatrics Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | - Domenico Accili
- Department of Medicine and Columbia University, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
| | | |
Collapse
|
28
|
Bahl AS, Verma VK, Bhatia J, Arya DS. Integrating in silico and in vivo approach for investigating the role of polyherbal oil in prevention and treatment of COVID-19 infection. Chem Biol Interact 2022; 367:110179. [PMID: 36113631 PMCID: PMC9472470 DOI: 10.1016/j.cbi.2022.110179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022]
Abstract
Currently, there are no FDA approved antiviral drugs available to treat COVID-19 patients. Also, due to emergence of new SARS-CoV-2 variants, the protective efficacy of vaccines could be reduced, hence it is urgent to have alternative treatments for combating the SARS-CoV-2 infection. Since, there is a long-standing history of herbal medicine in the treatment of respiratory diseases. In the present study, we investigated two polyherbal oil blend viz. Sudarshan AV and Elixir AV (SAV and EAV) in inhibiting SARS-COV-2. From GC-MS analysis of polyherbal oils (SAV and EAV) a total of 11 active compounds were selected, on the basis of their abundance and activity. Further, from the molecular docking studies, we found an inhibitory effect of these compounds on viral envelope and membrane, spike proteins whilst an agonistic effect with human host receptor angiotensin-converting enzyme 2 (ACE2) implicating the crucial role of the individual compound in resistance of SARS-CoV-2. Since, the in-silico results suggest that polyherbal oil (SAV and EAV) contributes in preventing the entry of SARS-CoV-2 into the human body, we further investigated the efficacy of polyherbal formulated essential oil (FEO; SAV & EAV) in prevention and treatment of COVID-19 in hamster model. The male golden Syrian hamsters (n = 23) were divided into 5 groups i.e., Group 1: Control (n = 3); Group 2: Infected (n = 5); Group 3: Infected + Remdesivir (n = 5); Group 4: Infected + FEO (n = 5) and Group 5: Prophylactic FEO + Infected (n = 5). In both treatment and prophylactic groups, the FEO's significantly reduced the lung injury investigated histo-pathologically and viral load expression measured by real time PCR in comparison to infected hamsters. Furthermore, cytokines expression analysis clearly highlighted the efficacy of FEO's due to its anti-inflammatory activity and overall protection in treatment groups. In conclusion, the FEO (SAV & EAV) seem to be potent in both prevention and treatment of COVID-19 and related lung injury.
Collapse
Affiliation(s)
- Amul S Bahl
- Department of Research, Development and Innovation, God's Own Store LLP, Delhi, India.
| | - Vipin Kumar Verma
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Jagriti Bhatia
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Dharamvir Singh Arya
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
29
|
Rizvi ZA, Babele P, Sadhu S, Madan U, Tripathy MR, Goswami S, Mani S, Kumar S, Awasthi A, Dikshit M. Prophylactic treatment of Glycyrrhiza glabra mitigates COVID-19 pathology through inhibition of pro-inflammatory cytokines in the hamster model and NETosis. Front Immunol 2022; 13:945583. [PMID: 36238303 PMCID: PMC9550929 DOI: 10.3389/fimmu.2022.945583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 09/05/2022] [Indexed: 01/08/2023] Open
Abstract
Severe coronavirus disease (COVID-19) is accompanied by acute respiratory distress syndrome and pulmonary pathology, and is presented mostly with an inflammatory cytokine release, a dysregulated immune response, a skewed neutrophil/lymphocyte ratio, and a hypercoagulable state. Though vaccinations have proved effective in reducing the COVID-19-related mortality, the limitation of the use of vaccine against immunocompromised individuals, those with comorbidity, and emerging variants remains a concern. In the current study, we investigate for the first time the efficacy of the Glycyrrhiza glabra (GG) extract, a potent immunomodulator, against SARS-CoV-2 infection in hamsters. Prophylactic treatment with GG showed protection against loss in body weight and a 35%–40% decrease in lung viral load along with reduced lung pathology in the hamster model. Remarkably, GG reduced the mRNA expression of pro-inflammatory cytokines and plasminogen activator inhibitor-1 (PAI-1). In vitro, GG acted as a potent immunomodulator by reducing Th2 and Th17 differentiation and IL-4 and IL-17A cytokine production. In addition, GG also showed robust potential to suppress ROS, mtROS, and NET generation in a concentration-dependent manner in both human polymorphonuclear neutrophils (PMNs) and murine bone marrow-derived neutrophils (BMDNs). Taken together, we provide evidence for the protective efficacy of GG against COVID-19 and its putative mechanistic insight through its immunomodulatory properties. Our study provides the proof of concept for GG efficacy against SARS-CoV-2 using a hamster model and opens the path for further studies aimed at identifying the active ingredients of GG and its efficacy in COVID-19 clinical cases.
Collapse
Affiliation(s)
- Zaigham Abbas Rizvi
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Prabhakar Babele
- Noncommunicable Disease Centre, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Srikanth Sadhu
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Upasna Madan
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Manas Ranjan Tripathy
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Sandeep Goswami
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Shailendra Mani
- Noncommunicable Disease Centre, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Sachin Kumar
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Amit Awasthi
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- *Correspondence: Madhu Dikshit, ; ; Amit Awasthi,
| | - Madhu Dikshit
- Noncommunicable Disease Centre, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- *Correspondence: Madhu Dikshit, ; ; Amit Awasthi,
| |
Collapse
|
30
|
Chen L, Wang S, Wang Z, Liu Y, Xu Y, Yang S, Xue G. Construction and analysis of competing endogenous RNA network and patterns of immune infiltration in abdominal aortic aneurysm. Front Cardiovasc Med 2022; 9:955838. [PMID: 35990982 PMCID: PMC9386163 DOI: 10.3389/fcvm.2022.955838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/12/2022] [Indexed: 11/29/2022] Open
Abstract
Background Various studies have highlighted the role of circular RNAs (circRNAs) as critical molecular regulators in cardiovascular diseases, but its role in abdominal aortic aneurysm (AAA) is unclear. This study explores the potential molecular mechanisms of AAA based on the circRNA-microRNA (miRNA)-mRNA competing endogenous RNA (ceRNA) network and immune cell infiltration patterns. Methods The expression profiles of circRNAs (GSE144431) and mRNAs (GSE57691 and GSE47472) were obtained from the Gene Expression Omnibus (GEO). Then, the differentially expressed circRNAs (DEcircRNAs) and mRNAs (DEmRNAs) between AAA patients and healthy control samples, and the target miRNAs of these DEmRNAs and DEcircRNAs were identified. Based on the miRNA-DEmRNAs and miRNA-DEcircRNAs pairs, the ceRNA network was constructed. Furthermore, the proportion of the 22 immune cell types in AAA patients was assessed using cell type identification by estimating relative subsets of RNA transcripts (CIBERSORT) algorithm. The expressions of key genes and immune cell infiltration were validated using clinical specimens. Results A total of 214 DEmRNAs were identified in the GSE57691 and GSE47472 datasets, and 517 DEcircRNAs were identified in the GSE144431 dataset. The ceRNA network included 19 circRNAs, 36 mRNAs, and 68 miRNAs. Two key genes, PPARG and FOXO1, were identified among the hub genes of the established protein-protein interaction between mRNAs in the ceRNA network. Moreover, seven types of immune cells were differentially expressed between AAA patients and healthy control samples. Hub genes in ceRNA, such as FOXO1, HSPA8, and RAB5C, positively correlated with resting CD4 memory T cells or M1 macrophages, or both. Conclusion In conclusion, a ceRNA interaction axis was constructed. The composition of infiltrating immune cells was analyzed in the abdominal aorta of AAA patients and healthy control samples. This may help identify potential therapeutic targets for AAA.
Collapse
Affiliation(s)
- Liang Chen
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shuangshuang Wang
- Songyuan Central Hospital, Songyuan Children's Hospital, Songyuan, China
| | - Zheyu Wang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yuting Liu
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yi Xu
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- *Correspondence: Shuofei Yang
| | - Guanhua Xue
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- Guanhua Xue
| |
Collapse
|
31
|
Ren X, Chen H. Changes in Th9 and Th17 lymphocytes and functional cytokines and their relationship with thyroid-stimulating hormone receptor antibodies at different stages of graves’ disease. Front Immunol 2022; 13:919681. [PMID: 35935994 PMCID: PMC9352852 DOI: 10.3389/fimmu.2022.919681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Graves’ disease (GD) is an organ-specific autoimmune disease characterized by the production of thyroid-stimulating antibodies (TSAb). The newly discovered CD4+ T helper cells, Th9 and Th17 lymphocytes, have been confirmed to be closely associated with a variety of immune diseases. However, relationships with the onset and development of GD remain unclear. The purpose of this study was to investigate the roles of Th9 and Th17 in the pathogenesis and prognosis of GD. Patients We recruited 26 patients with newly diagnosed GD, 45 patients with GD in remission, and 20 healthy individuals. Measurements Thyroid function and autoantibodies were evaluated using chemiluminescence immunoassays. Th9 and Th17 cells were analyzed using flow cytometry. The expression of Foxo1, IRF-4, RORc, IL-9, and IL-17 mRNA was examined using real-time PCR, and IL-9 and IL-17 protein levels were measured using enzyme-linked immunosorbent assay. Results Th9, Th17, and characteristic cytokines IL-9 and IL-17 in the GD-untreated group were significantly higher than those in the control and remission groups. The above indexes significantly decreased in the remission group, with the levels in the TRAb− remission group being similar to those in the normal group, while in the TRAb+ remission group, levels were differentially increased. TRAb titer was positively correlated with the levels of Th9, Th17, and their functional cytokines. Conclusions Th9 and Th17 cells may be involved in the pathogenesis and disease outcome of GD, which could provide a new direction for developing immunotherapy for patients with GD.
Collapse
|
32
|
Moon JH, Lee S, Pak M, Hur B, Kim S. MLDEG: A Machine Learning Approach to Identify Differentially Expressed Genes Using Network Property and Network Propagation. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2356-2364. [PMID: 33750713 DOI: 10.1109/tcbb.2021.3067613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
MOTIVATION Identifying differentially expressed genes (DEGs) in transcriptome data is a very important task. However, performances of existing DEG methods vary significantly for data sets measured in different conditions and no single statistical or machine learning model for DEG detection perform consistently well for data sets of different traits. In addition, setting a cutoff value for the significance of differential expressions is one of confounding factors to determine DEGs. RESULTS We address these problems by developing an ensemble model that refines the heterogeneous and inconsistent results of the existing methods by taking accounts into network information such as network propagation and network property. DEG candidates that are predicted with weak evidence by the existing tools are re-classified by our proposed ensemble model for the transcriptome data. Tested on 10 RNA-seq datasets downloaded from gene expression omnibus (GEO), our method showed excellent performance of winning the first place in detecting ground truth (GT) genes in eight datasets and find almost all GT genes in six datasets. On the other hand, performances of all existing methods varied significantly for the 10 data sets. Because of the design principle, our method can accommodate any new DEG methods naturally. AVAILABILITY The source code of our method is available at https://github.com/jihmoon/MLDEG.
Collapse
|
33
|
Zhang Q, Zhao J, Ni M, Shen Q, Zhou W, Liu Z. Vitamin D 3 reverses the transcriptional profile of offspring CD4 + T lymphocytes exposed to intrauterine inflammation. J Steroid Biochem Mol Biol 2022; 221:106120. [PMID: 35533917 DOI: 10.1016/j.jsbmb.2022.106120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/23/2022] [Accepted: 04/23/2022] [Indexed: 12/12/2022]
Abstract
Chorioamnionitis profoundly influences multiple fetal organs as well as the immune system. Maternal vitamin D (VitD) supplementation may modulate the immune function of offspring. Here, we sought to uncover the immunomodulatory potential of intrauterine inflammation and VitD in offspring CD4+ T cells. Pregnant C57BL/6 mice were treated with intrauterine lipopolysaccharide (LPS) injections, with or without VitD. Splenic CD4+ T cells were negatively selected using anti-biotin microbeads at 28 days after birth. Differentially expressed genes (DEGs) in the offspring CD4+ T cells were identified via RNA sequencing. In total, 181 DEGs induced by LPS exposure were identified in offspring CD4+ T cells. Furthermore, 2461 DEGs were detected after VitD supplementation in addition to LPS exposure. VitD supplementation showed an unexpected ability to counteract the LPS-induced transcriptional responses. VitD supplementation downregulated lymphocyte differentiation (GO: 0030098) and lymphocyte activation (GO: 0046649), and upregulated the responses to viruses (GO: 0009615) and bacteria (GO:0009617) in offspring CD4+ T cells with intrauterine LPS exposure. In addition, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that several pathways, including the T cell receptor signaling pathway, the mitogen-activated protein kinase (MAPK) signaling pathway, Th17 cell differentiation, and autophagy, were downregulated by intrauterine VitD intervention following LPS exposure. Subsequently, we confirmed the counteracting effect of VitD against LPS on the expression of several genes (Insr, Foxo1, and Peli1) using qRT-PCR and western blot analyses. We also demonstrated that intrauterine VitD supplementation interferes with offspring Th17 cell differentiation induced by intrauterine LPS exposure. Our study revealed that VitD reverses the transcriptional and Th17 differential profiles of offspring CD4+ T lymphocytes induced by intrauterine LPS, and indicated the contribution of maternal VitD supplementation to immune protection in offspring affected by intrauterine inflammation.
Collapse
Affiliation(s)
- Qianqian Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China; International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Jiuru Zhao
- International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Meng Ni
- International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Qianwen Shen
- International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Wenhao Zhou
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai, China.
| | - Zhiwei Liu
- International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
34
|
Machida K, Tahara SM. Immunotherapy and Microbiota for Targeting of Liver Tumor-Initiating Stem-like Cells. Cancers (Basel) 2022; 14:2381. [PMID: 35625986 PMCID: PMC9139909 DOI: 10.3390/cancers14102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 02/08/2023] Open
Abstract
Cancer contains tumor-initiating stem-like cells (TICs) that are resistant to therapies. Hepatocellular carcinoma (HCC) incidence has increased twice over the past few decades, while the incidence of other cancer types has trended downward globally. Therefore, an understanding of HCC development and therapy resistance mechanisms is needed for this incurable malignancy. This review article describes links between immunotherapies and microbiota in tumor-initiating stem-like cells (TICs), which have stem cell characteristics with self-renewal ability and express pluripotency transcription factors such as NANOG, SOX2, and OCT4. This review discusses (1) how immunotherapies fail and (2) how gut dysbiosis inhibits immunotherapy efficacy. Gut dysbiosis promotes resistance to immunotherapies by breaking gut immune tolerance and activating suppressor immune cells. Unfortunately, this leads to incurable recurrence/metastasis development. Personalized medicine approaches targeting these mechanisms of TIC/metastasis-initiating cells are emerging targets for HCC immunotherapy and microbiota modulation therapy.
Collapse
Affiliation(s)
- Keigo Machida
- Southern California Research Center for ALPD and Cirrhosis, Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 2011 Zonal Ave., 503C-HMR, Los Angeles, CA 90033, USA;
| | | |
Collapse
|
35
|
Musiol S, Alessandrini F, Jakwerth CA, Chaker AM, Schneider E, Guerth F, Schnautz B, Grosch J, Ghiordanescu I, Ullmann JT, Kau J, Plaschke M, Haak S, Buch T, Schmidt-Weber CB, Zissler UM. TGF-β1 Drives Inflammatory Th Cell But Not Treg Cell Compartment Upon Allergen Exposure. Front Immunol 2022; 12:763243. [PMID: 35069535 PMCID: PMC8777012 DOI: 10.3389/fimmu.2021.763243] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
TGF-β1 is known to have a pro-inflammatory impact by inducing Th9 and Th17 cells, while it also induces anti-inflammatory Treg cells (Tregs). In the context of allergic airway inflammation (AAI) its dual role can be of critical importance in influencing the outcome of the disease. Here we demonstrate that TGF-β is a major player in AAI by driving effector T cells, while Tregs differentiate independently. Induction of experimental AAI and airway hyperreactivity in a mouse model with inducible genetic ablation of the gene encoding for TGFβ-receptor 2 (Tgfbr2) on CD4+T cells significantly reduced the disease phenotype. Further, it blocked the induction of pro-inflammatory T cell frequencies (Th2, Th9, Th17), but increased Treg cells. To translate these findings into a human clinically relevant context, Th2, Th9 and Treg cells were quantified both locally in induced sputum and systemically in blood of allergic rhinitis and asthma patients with or without allergen-specific immunotherapy (AIT). Natural allergen exposure induced local and systemic Th2, Th9, and reduced Tregs cells, while therapeutic allergen exposure by AIT suppressed Th2 and Th9 cell frequencies along with TGF-β and IL-9 secretion. Altogether, these findings support that neutralization of TGF-β represents a viable therapeutic option in allergy and asthma, not posing the risk of immune dysregulation by impacting Tregs cells.
Collapse
Affiliation(s)
- Stephanie Musiol
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Francesca Alessandrini
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Constanze A Jakwerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Adam M Chaker
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany.,Department of Otorhinolaryngology, Klinikum rechts der Isar, TUM School of Medicine, Technical University Munich, Munich, Germany
| | - Evelyn Schneider
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Ferdinand Guerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Benjamin Schnautz
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Johanna Grosch
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Ileana Ghiordanescu
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Julia T Ullmann
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Josephine Kau
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Mirjam Plaschke
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Stefan Haak
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Carsten B Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Ulrich M Zissler
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| |
Collapse
|
36
|
Rizvi ZA, Dalal R, Sadhu S, Binayke A, Dandotiya J, Kumar Y, Shrivastava T, Gupta SK, Aggarwal S, Tripathy MR, Rathore DK, Yadav AK, Medigeshi GR, Pandey AK, Samal S, Asthana S, Awasthi A. Golden Syrian hamster as a model to study cardiovascular complications associated with SARS-CoV-2 infection. eLife 2022; 11:73522. [PMID: 35014610 PMCID: PMC8794466 DOI: 10.7554/elife.73522] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 infection in the Golden Syrian hamster causes lung pathology that resembles human coronavirus disease (COVID-19). However, extra-pulmonary pathologies associated with SARS-CoV-2 infection and post COVID sequelae remain to be understood. Here we show, using a hamster model, that the early phase of SARS-CoV-2 infection leads to an acute inflammatory response and lung pathologies, while the late phase of infection causes cardiovascular complications (CVC) characterized by ventricular wall thickening associated with increased ventricular mass/ body mass ratio and interstitial coronary fibrosis. Molecular profiling further substantiated our findings of CVC, as SARS-CoV-2-infected hamsters showed elevated levels of serum cardiac Troponin-I (cTnI), cholesterol, low-density lipoprotein and long-chain fatty acid triglycerides. Serum metabolomics profiling of SARS-CoV-2-infected hamsters identified N-acetylneuraminate, a functional metabolite found to be associated with CVC, as a metabolic marker was found to be common between SARS-CoV-2-infected hamsters and COVID-19 patients. Together, we propose hamsters as a suitable animal model to study post-COVID sequelae associated with CVC which could be extended to therapeutic interventions.
Collapse
Affiliation(s)
- Zaigham Abbas Rizvi
- Immuno-biology Lab, Infection and Immunology centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Rajdeep Dalal
- Immuno-biology Lab, Infection and Immunology centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Srikanth Sadhu
- Immuno-biology Lab, Infection and Immunology centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Akshay Binayke
- Immuno-biology Lab, Infection and Immunology centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Jyotsna Dandotiya
- Immuno-biology Lab, Infection and Immunology centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Yashwant Kumar
- Non-communicable disease centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Tripti Shrivastava
- Infection and Immunology centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Sonu Kumar Gupta
- Non-communicable disease centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Suruchi Aggarwal
- Non-communicable disease centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Manas Ranjan Tripathy
- Immuno-biology Lab, Infection and Immunology centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Deepak Kumar Rathore
- Infection and Immunology Center, Translational Health Science and Technology Institute, Faridabad, India
| | - Amit Kumar Yadav
- Non-communicable disease center, Translational Health Science and Technology Institute, Faridabad, India
| | - Guruprasad R Medigeshi
- Infection and Immunology Center, Translational Health Science and Technology Institute, Gurgaon, India
| | - Amit Kumar Pandey
- Infection and Immunology Center, Translational Health Science and Technology Institute, Faridabad, India
| | - Sweety Samal
- Infection and Immunology Center, Translational Health Science and Technology Institute, Faridabad, India
| | - Shailendra Asthana
- Non-communicable disease centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Amit Awasthi
- Immuno-biology Lab, Infection and Immunology centre, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
37
|
Cameron B, Zaheer SA, Dominguez-Villar M. Control of CD4+ T Cell Differentiation and Function by PI3K Isoforms. Curr Top Microbiol Immunol 2022; 436:197-216. [DOI: 10.1007/978-3-031-06566-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
38
|
Rizvi ZA, Tripathy MR, Sharma N, Goswami S, Srikanth N, Sastry JLN, Mani S, Surjit M, Awasthi A, Dikshit M. Effect of Prophylactic Use of Intranasal Oil Formulations in the Hamster Model of COVID-19. Front Pharmacol 2021; 12:746729. [PMID: 34721035 PMCID: PMC8551705 DOI: 10.3389/fphar.2021.746729] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/10/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection initiates with viral entry in the upper respiratory tract, leading to coronavirus disease 2019 (COVID-19). Severe COVID-19 is characterized by pulmonary pathologies associated with respiratory failure. Thus, therapeutics aimed at inhibiting the entry of the virus or its internalization in the upper respiratory tract are of interest. Herein, we report the prophylactic application of two intranasal formulations provided by the National Medicinal Plant Board (NMPB), Anu oil and til tailya, in the hamster model of SARS-CoV-2 infection. Prophylactic intra-nasal instillation of these oil formulations exhibited reduced viral load in lungs and resulted in reduced body weight loss and lung-pneumonitis. In line with reduced viral load, histopathological analysis revealed a reduction in lung pathology in the Anu oil group as compared to the control infected group. However, the til tailya group did not show a significant reduction in lung pathology. Furthermore, molecular analysis using mRNA expression profiling indicated reduced expression of pro-inflammatory cytokine genes, including Th1 and Th17 cytokines for both the intranasal formulations as a result of decreased viral load. Together, the prophylactic intranasal application of Anu oil seems to be useful in limiting both viral load and severity in SARS-CoV2 infection in the hamster model.
Collapse
Affiliation(s)
- Zaigham Abbas Rizvi
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, India
| | - Manas Ranjan Tripathy
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, India
| | - Nishant Sharma
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, India
| | - Sandeep Goswami
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, India
| | - N Srikanth
- DG(I/C), Central Council for Ayurvedic Sciences, New Delhi, India
| | - J L N Sastry
- CEO-National Medicinal Plants Board, Ministry of AYUSH, New Delhi, India
| | - Shailendra Mani
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, India
| | - Milan Surjit
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, India
| | - Amit Awasthi
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, India
| | - Madhu Dikshit
- Non-communicable Disease Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, India
| |
Collapse
|
39
|
D'Souza S, Nair AP, Sahu GR, Vaidya T, Shetty R, Khamar P, Mullick R, Gupta S, Dickman MM, Nuijts RMMA, Mohan RR, Ghosh A, Sethu S. Keratoconus patients exhibit a distinct ocular surface immune cell and inflammatory profile. Sci Rep 2021; 11:20891. [PMID: 34686755 PMCID: PMC8536707 DOI: 10.1038/s41598-021-99805-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammatory factors have been considered to contribute to keratoconus (KC) pathogenesis. This study aims to determine the immune cells subsets and soluble inflammatory factor profile on the ocular surface of KC patients. 32 KC subjects (51 eyes) across different grades of severity and 15 healthy controls (23 eyes) were included in the study. Keratometry and pachymetry measurements were recorded. Ocular surface immune cells (collected by ocular surface wash) immunophenotyped using flow cytometry include leukocytes, neutrophils, macrophages, natural killer (NK) cells, pan-T cells, gamma delta T (γδT) cells and NKT cells. Tear fluid collected using Schirmer's strip was used to measure 50 soluble factors by multiplex ELISA. Proportions of activated neutrophils, NK cells and γδT cells were significantly increased in KC patients. Significantly higher levels of tear fluid IL-1β, IL-6, LIF, IL-17A, TNFα, IFNα/β/γ, EPO, TGFβ1, PDGF-BB, sVCAM, sL-selectin, granzyme-B, perforin, MMP2, sFasL and IgE, along with significantly lower levels of IL-1α and IL-9 were observed in KC patients. Alterations observed in few of the immuno-inflammatory parameters correlated with grades of disease, allergy, eye rubbing and keratometry or pachymetry measurements. The observation implies a distinct immuno-inflammatory component in KC pathogenesis and its potential as an additional therapeutic target in KC management.
Collapse
Affiliation(s)
- Sharon D'Souza
- Department of Cornea and Refractive Surgery, Narayana Nethralaya, Bangalore, India
| | - Archana Padmanabhan Nair
- GROW Research Laboratory, Narayana Nethralaya Foundation, 3rd Floor, Narayana Nethralaya, #258/A Hosur Road, Bommasandra, Bangalore, 560099, India.,Manipal Academy of Higher Education, Manipal, India
| | - Ganesh Ram Sahu
- GROW Research Laboratory, Narayana Nethralaya Foundation, 3rd Floor, Narayana Nethralaya, #258/A Hosur Road, Bommasandra, Bangalore, 560099, India
| | - Tanuja Vaidya
- GROW Research Laboratory, Narayana Nethralaya Foundation, 3rd Floor, Narayana Nethralaya, #258/A Hosur Road, Bommasandra, Bangalore, 560099, India.,Manipal Academy of Higher Education, Manipal, India
| | - Rohit Shetty
- Department of Cornea and Refractive Surgery, Narayana Nethralaya, Bangalore, India
| | - Pooja Khamar
- Department of Cornea and Refractive Surgery, Narayana Nethralaya, Bangalore, India
| | - Ritika Mullick
- Department of Cornea and Refractive Surgery, Narayana Nethralaya, Bangalore, India
| | - Sneha Gupta
- Department of Cornea and Refractive Surgery, Narayana Nethralaya, Bangalore, India
| | - Mor M Dickman
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands.,MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Rudy M M A Nuijts
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Rajiv R Mohan
- Department of Veterinary Medicine and Surgery, University of Missouri, 1600 E. Rollins Rd, Columbia, MO, 65211, USA. .,Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO, USA. .,Harry S Truman Veterans' Memorial Hospital, Columbia, MO, USA.
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, 3rd Floor, Narayana Nethralaya, #258/A Hosur Road, Bommasandra, Bangalore, 560099, India. .,Singapore Eye Research Institute, Singapore, Singapore.
| | - Swaminathan Sethu
- GROW Research Laboratory, Narayana Nethralaya Foundation, 3rd Floor, Narayana Nethralaya, #258/A Hosur Road, Bommasandra, Bangalore, 560099, India.
| |
Collapse
|
40
|
Wang RK, Sun YY, Li GY, Yang HT, Liu XJ, Li KF, Zhu X, Yu GY. MicroRNA-124-5p delays the progression of cerebral aneurysm by regulating FoxO1. Exp Ther Med 2021; 22:1172. [PMID: 34504617 PMCID: PMC8393823 DOI: 10.3892/etm.2021.10606] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 03/17/2021] [Indexed: 12/17/2022] Open
Abstract
Cerebral aneurysm (CA) is a common brain disease, and the development of cerebral aneurysm is driven by inflammation and hemodynamic stress. MicroRNA (miR)-124-5p is reported to be associated with inflammatory response in brain disease such as cerebral ischemia-reperfusion injury. However, the function and molecular mechanism of miR-124-5p in CA are not clear, thus, the effects of miR-124-5p on inflammatory response in CA were explored. Firstly, the expression of miR-124-5p in the peripheral blood of patients with CA and the control group was detected by reverse transcription-quantitative PCR. Then, the human umbilical vein endothelial cells (HUVECs) were used as an in vitro model system and stimulated with interleukin (IL)-1β to simulate the inflammatory environment of CA, and the expression of miR-124-5p was detected. Next, the effect of miR-124-5p on the migration and invasion of HUVECs was detected using Transwell assays. Meanwhile, the function of miR-124-5p on various inflammatory factors was determined by western blotting and enzyme-linked immunosorbent assay (ELISA). Next, the TargetScan website was used to predict FoxO1 as a target gene of miR-124-5p, and this target association was validated by double luciferase reporter assay and western blotting. Finally, the interaction of miR-124-5p with FoxO1 in CA was measured by Transwell western blotting and ELISA assays. The results showed that the expression level of miR-124-5p in the peripheral blood of patients with CA was lower compared with that of control group, and the miR-124-5p in HUVECs stimulated by IL-1β was less compared with that in normal HUVECs. Besides, miR-124-5p could inhibit the migration and invasion abilities of HUVECs and the release of inflammatory factors. Additionally, the overexpression of miR-124-5p was able to inhibit the expression of FoxO1. miR-124-5p-inhibitor promoted the migration and invasion of HUVECs, as well as inflammatory response, which was weakened following the introduction of FoxO1 small interfering RNA. Overall, the present study demonstrated that miR-124-5p could prevent the occurrence and development of cerebral aneurysm by downregulating the expression of FoxO1.
Collapse
Affiliation(s)
- Ru-Ke Wang
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Yuan-Yuan Sun
- CT Room, Handan First Hospital, Handan, Hebei 056002, P.R. China
| | - Guang-You Li
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Hua-Tang Yang
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Xiu-Jie Liu
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Ke-Feng Li
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Xu Zhu
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Guo-Yuan Yu
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| |
Collapse
|
41
|
Rizvi ZA, Dalal R, Sadhu S, Kumar Y, Kumar S, Gupta SK, Tripathy MR, Rathore DK, Awasthi A. High-salt diet mediates interplay between NK cells and gut microbiota to induce potent tumor immunity. SCIENCE ADVANCES 2021; 7:eabg5016. [PMID: 34516769 PMCID: PMC8442882 DOI: 10.1126/sciadv.abg5016] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
High-salt diet (HSD) modulates effector and regulatory T cell functions and promotes tissue inflammation in autoimmune diseases. However, effects of HSD and its association with gut microbiota in tumor immunity remain undefined. Here, we report that HSD induces natural killer (NK) cell–mediated tumor immunity by inhibiting PD-1 expression while enhancing IFNγ and serum hippurate. Salt enhanced tumor immunity when combined with a suboptimal dose of anti-PD1 antibody. While HSD-induced tumor immunity was blunted upon gut microbiota depletion, fecal microbiota transplantation (FMT) from HSD mice restored the tumor immunity associated with NK cell functions. HSD increased the abundance of Bifidobacterium and caused increased gut permeability leading to intratumor localization of Bifidobacterium, which enhanced NK cell functions and tumor regression. Intratumoral injections of Bifidobacterium activated NK cells, which inhibited tumor growth. These results indicate that HSD modulates gut microbiome that induces NK cell–dependent tumor immunity with a potential translational perspective.
Collapse
Affiliation(s)
- Zaigham Abbas Rizvi
- Immunbiology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Infection and Immunology, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Rajdeep Dalal
- Immunbiology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Infection and Immunology, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Srikanth Sadhu
- Immunbiology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Infection and Immunology, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Yashwant Kumar
- Noncommunicable Disease Center, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Shakti Kumar
- Infection and Immunology, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Sonu Kumar Gupta
- Noncommunicable Disease Center, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Manas Ranjan Tripathy
- Immunbiology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Infection and Immunology, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Deepak Kumar Rathore
- Infection and Immunology, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Amit Awasthi
- Immunbiology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Infection and Immunology, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Corresponding author.
| |
Collapse
|
42
|
Schanz O, Cornez I, Yajnanarayana SP, David FS, Peer S, Gruber T, Krawitz P, Brossart P, Heine A, Landsberg J, Baier G, Wolf D. Tumor rejection in Cblb -/- mice depends on IL-9 and Th9 cells. J Immunother Cancer 2021; 9:jitc-2021-002889. [PMID: 34272310 PMCID: PMC8287598 DOI: 10.1136/jitc-2021-002889] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Casitas B lymphoma-b (Cbl-b) is a central negative regulator of cytotoxic T and natural killer (NK) cells and functions as an intracellular checkpoint in cancer. In particular, Th9 cells support mast cell activation, promote dendritic cell recruitment, enhance the cytolytic function of cytotoxic T lymphocytes and NK cells, and directly kill tumor cells, thereby contributing to tumor immunity. However, the role of Cbl-b in the differentiation and antitumor function of Th9 cells is not sufficiently resolved. METHODS Using Cblb -/- mice, we investigated the effect of knocking out Cblb on the differentiation process and function of different T helper cell subsets, focusing on regulatory T cell (Treg) and Th9 cells. We applied single-cell RNA (scRNA) sequencing of in vitro differentiated Th9 cells to understand how Cbl-b shapes the transcriptome and regulates the differentiation and function of Th9 cells. We transferred tumor-model antigen-specific Cblb -/- Th9 cells into melanoma-bearing mice and assessed tumor control in vivo. In addition, we blocked interleukin (IL)-9 in melanoma cell-exposed Cblb -/- mice to investigate the role of IL-9 in tumor immunity. RESULTS Here, we provide experimental evidence that Cbl-b acts as a rheostat favoring Tregs at the expense of Th9 cell differentiation. Cblb -/- Th9 cells exert superior antitumor activity leading to improved melanoma control in vivo. Accordingly, blocking IL-9 in melanoma cell-exposed Cblb -/- mice reversed their tumor rejection phenotype. Furthermore, scRNA sequencing of in vitro differentiated Th9 cells from naïve T cells isolated from wildtype and Cblb -/- animals revealed a transcriptomic basis for increased Th9 cell differentiation. CONCLUSION We established IL-9 and Th9 cells as key antitumor executers in Cblb -/- animals. This knowledge may be helpful for the future improvement of adoptive T cell therapies in cancer.
Collapse
Affiliation(s)
- Oliver Schanz
- Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Isabelle Cornez
- Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | | | - Friederike Sophie David
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Bonn, Germany.,Institute of Human Genetics, University Hospital Bonn, Bonn, Germany
| | - Sebastian Peer
- Institute of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Gruber
- Institute of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Bonn, Germany
| | - Peter Brossart
- Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Annkristin Heine
- Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | | | - Gottfried Baier
- Institute of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Dominik Wolf
- Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany .,Department of Internal Medicine V, Hematology and Oncology, and Tyrolean Cancer Research Institute (TKFI), Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
43
|
Diversity of T Helper and Regulatory T Cells and Their Contribution to the Pathogenesis of Allergic Diseases. Handb Exp Pharmacol 2021; 268:265-296. [PMID: 34247282 DOI: 10.1007/164_2021_486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
T helper (Th) and regulatory T (Treg) cells represent important effectors of adaptive immunity. They mediate communication between the immune system and tissue sites and thereby coordinate effective defense against environmental threats or maintain tolerance, respectively. Since the discovery of two prototypic T helper cells, Th1 and Th2, additional phenotypic and functional distinct subsets have been described ranging from Th17, Th22, Th9, and T follicular helper cells. The same holds true for regulatory T cells that represent a family with functionally distinct subsets characterized by co-expression of the transcription factors T-bet, Gata3, or RORγt. Here, we summarize the current knowledge on differentiation and function of T helper and regulatory T cell subsets and discuss their lineage stability versus plasticity towards other subsets. In addition, we highlight the direct and indirect contribution of each subset to the pathology of allergies and indicate novel therapies for specific targeting the effector functions of T helper and regulatory T cells.
Collapse
|
44
|
Cheema PS, Nandi D, Nag A. Exploring the therapeutic potential of forkhead box O for outfoxing COVID-19. Open Biol 2021; 11:210069. [PMID: 34102081 PMCID: PMC8187014 DOI: 10.1098/rsob.210069] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/27/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic has wreaked unprecedented societal havoc worldwide. The infected individuals may present mild to severe symptoms, with nearly 20% of the confirmed patients impaired with significant complications, including multi-organ failure. Acute respiratory distress imposed by SARS-CoV-2 largely results from an aggravated cytokine storm and deregulated immune response. The forkhead box O (FoxO) transcription factors are reported to play a significant role in maintaining normal cell physiology by regulating survival, apoptosis, oxidative stress, development and maturation of T and B lymphocytes, secretion of inflammatory cytokines, etc. We propose a potent anti-inflammatory approach based on activation of the FoxO as an attractive strategy against the novel coronavirus. This regime will be focused on restoring redox and inflammatory homeostasis along with repair of the damaged tissue, activation of lymphocyte effector and memory cells. Repurposing FoxO activators as a means to alleviate the inflammatory burst following SARS-CoV-2 infection can prove immensely valuable in the ongoing pandemic and provide a reliable groundwork for enriching our repertoire of antiviral modalities for any such complication in the future. Altogether, our review highlights the possible efficacy of FoxO activation as a novel arsenal for clinical management of COVID-19.
Collapse
Affiliation(s)
- Pradeep Singh Cheema
- Department of Biochemistry, University of Delhi, South Campus, Biotech Building, 2nd Floor, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| | - Deeptashree Nandi
- Department of Biochemistry, University of Delhi, South Campus, Biotech Building, 2nd Floor, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi, South Campus, Biotech Building, 2nd Floor, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| |
Collapse
|
45
|
Enhanced IL-9 secretion by p66Shc-deficient CLL cells modulates the chemokine landscape of the stromal microenvironment. Blood 2021; 137:2182-2195. [PMID: 33181836 DOI: 10.1182/blood.2020005785] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/04/2020] [Indexed: 12/19/2022] Open
Abstract
The stromal microenvironment is central to chronic lymphocytic leukemia (CLL) pathogenesis. How leukemic cells condition the stroma to enhance its chemoattractant properties remains elusive. Here, we show that mouse and human CLL cells promote the contact-independent stromal expression of homing chemokines. This function was strongly enhanced in leukemic cells from Eμ-TCL1 mice lacking the pro-oxidant p66Shc adaptor, which develop an aggressive disease with organ infiltration. We identified interleukin-9 (IL-9) as the soluble factor, negatively modulated by p66Shc, that is responsible for the chemokine-elevating activity of leukemic cells on stromal cells. IL-9 blockade in Eμ-TCL1/p66Shc-/- mice resulted in a decrease in the nodal expression of homing chemokines, which correlated with decreased leukemic cell invasiveness. IL-9 levels were found to correlate inversely with residual p66Shc in p66Shc-deficient human CLL cells (n = 52 patients). p66Shc reconstitution in CLL cells normalized IL-9 expression and neutralized their chemokine-elevating activity. Notably, high IL-9 expression in CLL cells directly correlates with lymphadenopathy, liver infiltration, disease severity, and overall survival, emerging as an independent predictor of disease outcome. Our results demonstrate that IL-9 modulates the chemokine landscape in the stroma and that p66Shc, by regulating IL-9 expression, fine tunes the ability of leukemic cells to shape the microenvironment, thereby contributing to CLL pathogenesis.
Collapse
|
46
|
Abstract
CD4 T cell effector subsets not only profoundly affect cancer progression, but recent evidence also underscores their critical contribution to the anticancer efficacy of immune checkpoint inhibitors. In 2012, the two seminal studies suggested the superior antimelanoma activity of TH9 cells over other T cell subsets upon adoptive T cell transfer. While these findings provided great impetus to investigate further the unique functions of TH9 cells and explore their relevance in cancer immunotherapy, the following questions still remain outstanding: are TH9 cell anticancer functions restricted to melanoma? What are the factors favouring TH9 cell effector functions? What is the contribution of TH9 cells to cancer immunotherapy treatments? Can TH9 cells be identified in humans and, if so, what is their clinical relevance? By reviewing the studies addressing these questions, we will discuss how TH9 cells could be therapeutically harnessed for cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Isis Benoit-Lizon
- INSERM, U1231, Dijon, France; Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France
| | - Lionel Apetoh
- INSERM, U1231, Dijon, France; Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France; Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
47
|
Sadhu S, Rizvi ZA, Pandey RP, Dalal R, Rathore DK, Kumar B, Pandey M, Kumar Y, Goel R, Maiti TK, Johri AK, Tiwari A, Pandey AK, Awasthi A. Gefitinib Results in Robust Host-Directed Immunity Against Salmonella Infection Through Proteo-Metabolomic Reprogramming. Front Immunol 2021; 12:648710. [PMID: 33868285 PMCID: PMC8044459 DOI: 10.3389/fimmu.2021.648710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/08/2021] [Indexed: 11/17/2022] Open
Abstract
The global rise of antibiotic-resistant strains of Salmonella has necessitated the development of alternative therapeutic strategies. Recent studies have shown that targeting host factors may provide an alternative approach for the treatment of intracellular pathogens. Host-directed therapy (HDT) modulates host cellular factors that are essential to support the replication of the intracellular pathogens. In the current study, we identified Gefitinib as a potential host directed therapeutic drug against Salmonella. Further, using the proteome analysis of Salmonella-infected macrophages, we identified EGFR, a host factor, promoting intracellular survival of Salmonella via mTOR-HIF-1α axis. Blocking of EGFR, mTOR or HIF-1α inhibits the intracellular survival of Salmonella within the macrophages and in mice. Global proteo-metabolomics profiling indicated the upregulation of host factors predominantly associated with ATP turn over, glycolysis, urea cycle, which ultimately promote the activation of EGFR-HIF1α signaling upon infection. Importantly, inhibition of EGFR and HIF1α restored both proteomics and metabolomics changes caused by Salmonella infection. Taken together, this study identifies Gefitinib as a host directed drug that holds potential translational values against Salmonella infection and might be useful for the treatment of other intracellular infections.
Collapse
Affiliation(s)
- Srikanth Sadhu
- Infection and Immunobiology, Translational Health Science and Technology Institute, Faridabad, India
| | - Zaigham Abbas Rizvi
- Infection and Immunobiology, Translational Health Science and Technology Institute, Faridabad, India
| | | | - Rajdeep Dalal
- Infection and Immunobiology, Translational Health Science and Technology Institute, Faridabad, India
| | - Deepak Kumar Rathore
- Infection and Immunity, Translational Health Science and Technology Institute, Faridabad, India
| | - Bhoj Kumar
- Functional Proteomics Laboratory, Regional Centre for Biotechnology, Faridabad, India
| | - Manitosh Pandey
- Infection and Immunity, Translational Health Science and Technology Institute, Faridabad, India
| | - Yashwant Kumar
- Non Communicable Diseases, Translational Health Science and Technology Institute, Faridabad, India
| | - Renu Goel
- Non Communicable Diseases, Translational Health Science and Technology Institute, Faridabad, India
| | - Tushar K. Maiti
- Functional Proteomics Laboratory, Regional Centre for Biotechnology, Faridabad, India
| | - Atul Kumar Johri
- Infection and Immunity, Jawaharlal Nehru University, New Delhi, India
| | - Ashutosh Tiwari
- Infection and Immunobiology, Translational Health Science and Technology Institute, Faridabad, India
| | - Amit Kumar Pandey
- Infection and Immunity, Translational Health Science and Technology Institute, Faridabad, India
| | - Amit Awasthi
- Infection and Immunobiology, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
48
|
Critical Roles of Balanced T Helper 9 Cells and Regulatory T Cells in Allergic Airway Inflammation and Tumor Immunity. J Immunol Res 2021; 2021:8816055. [PMID: 33748292 PMCID: PMC7943311 DOI: 10.1155/2021/8816055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/12/2021] [Accepted: 02/17/2021] [Indexed: 01/02/2023] Open
Abstract
CD4+T helper (Th) cells are important mediators of immune responses in asthma and cancer. When counteracted by different classes of pathogens, naïve CD4+T cells undergo programmed differentiation into distinct types of Th cells. Th cells orchestrate antigen-specific immune responses upon their clonal T-cell receptor (TCR) interaction with the appropriate peptide antigen presented on MHC class II molecules expressed by antigen-presenting cells (APCs). T helper 9 (Th9) cells and regulatory T (Treg) cells and their corresponding cytokines have critical roles in tumor and allergic immunity. In the context of asthma and cancer, the dynamic internal microenvironment, along with chronic inflammatory stimuli, influences development, differentiation, and function of Th9 cells and Treg cells. Furthermore, the dysregulation of the balance between Th9 cells and Treg cells might trigger aberrant immune responses, resulting in development and exacerbation of asthma and cancer. In this review, the development, differentiation, and function of Th9 cells and Treg cells, which are synergistically regulated by various factors including cytokine signals, transcriptional factors (TFs), costimulatory signals, microenvironment cues, metabolic pathways, and different signal pathways, will be discussed. In addition, we focus on the recent progress that has helped to achieve a better understanding of the roles of Th9 cells and Treg cells in allergic airway inflammation and tumor immunity. We also discuss how various factors moderate their responses in asthma and cancer. Finally, we summarize the recent findings regarding potential mechanisms for regulating the balance between Th9 and Treg cells in asthma and cancer. These advances provide opportunities for novel therapeutic strategies that are aimed at reestablishing the balance of these cells in the diseases.
Collapse
|
49
|
Sharma D. Meet Our Editorial Board Member. Comb Chem High Throughput Screen 2020. [DOI: 10.2174/138620732309201127092910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Deepak Sharma
- Indian Institute of Technology Roorkee Roorkee 247667, India
| |
Collapse
|
50
|
Gang H, Peng D, Hu Y, Tang S, Li S, Huang Q. Interleukin-9-secreting CD4 + T cells regulate CD8 + T cells cytotoxicity in patients with acute coronary syndromes. APMIS 2020; 129:91-102. [PMID: 33113251 DOI: 10.1111/apm.13094] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/22/2020] [Indexed: 12/21/2022]
Abstract
T cells play vital roles in the development and progression of acute coronary syndromes (ACS), including cytotoxicity mediated by CD8+ T cells and immunoregulatory activity mediated by CD4+ T cells. Interleukin (IL)-9-secreting CD4+ T cells (Th9 cells) were recently found to be involved in the onset of ACS. We investigated regulatory role of Th9 cells to CD8+ T cells in patients with stable angina pectoris, unstable angina pectoris, and acute myocardial infarction (AMI). Circulating Th9 cells percentage, plasma IL-9 level, and PU.1 mRNA relative level was up-regulated in AMI patients compared with controls. There was no significant difference of IL-9-secreting CD8+ T cells percentage among groups. CD8+ T cells from AMI patients revealed increased cytotoxicity than those from controls, which presented as enhanced cytotolytic activity to target cells, increased interferon-γ and tumor necrosis factor-α secretion, elevated perforin and granzyme B production, and reduced programmed death-1 and cytotoxic T lymphocyte-associated protein 4. IL-9 stimulation did not affect proliferation, but promoted CD8+ T-cell cytotoxicity from both controls and AMI patients. IL-9-secreting CD4+ T cells were enriched in CD4+ CCR4- CCR6- CXCR3- cells. The enhancement of CD8+ T-cell cytotoxicity induced by CD4+ CCR4- CCR6- CXCR3- cells was dependent on IL-9 secretion. The present results indicated that up-regulation of IL-9-secreting CD4+ T cells may contribute to pathogenesis of AMI through enhancement of CD8+ T-cell cytotoxicity.
Collapse
Affiliation(s)
- Hongsheng Gang
- Department of Cardiology, Wuhan Fourth Hospital, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dingfeng Peng
- Department of Cardiology, Wuhan Fourth Hospital, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongjun Hu
- Department of Cardiology, Wuhan Fourth Hospital, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoyong Tang
- Department of Cardiology, Wuhan Fourth Hospital, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songhai Li
- Department of Cardiology, Wuhan Fourth Hospital, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Huang
- Department of Cardiology, Wuhan Fourth Hospital, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|