1
|
An Y, Ni R, Zhuang L, Yang L, Ye Z, Li L, Parkkila S, Aspatwar A, Gong W. Tuberculosis vaccines and therapeutic drug: challenges and future directions. MOLECULAR BIOMEDICINE 2025; 6:4. [PMID: 39841361 PMCID: PMC11754781 DOI: 10.1186/s43556-024-00243-6] [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: 08/21/2024] [Revised: 12/06/2024] [Accepted: 12/24/2024] [Indexed: 01/23/2025] Open
Abstract
Tuberculosis (TB) remains a prominent global health challenge, with the World Health Organization documenting over 1 million annual fatalities. Despite the deployment of the Bacille Calmette-Guérin (BCG) vaccine and available therapeutic agents, the escalation of drug-resistant Mycobacterium tuberculosis strains underscores the pressing need for more efficacious vaccines and treatments. This review meticulously maps out the contemporary landscape of TB vaccine development, with a focus on antigen identification, clinical trial progress, and the obstacles and future trajectories in vaccine research. We spotlight innovative approaches, such as multi-antigen vaccines and mRNA technology platforms. Furthermore, the review delves into current TB therapeutics, particularly for multidrug-resistant tuberculosis (MDR-TB), exploring promising agents like bedaquiline (BDQ) and delamanid (DLM), as well as the potential of host-directed therapies. The hurdles in TB vaccine and therapeutic development encompass overcoming antigen diversity, enhancing vaccine effectiveness across diverse populations, and advancing novel vaccine platforms. Future initiatives emphasize combinatorial strategies, the development of anti-TB compounds targeting novel pathways, and personalized medicine for TB treatment and prevention. Despite notable advances, persistent challenges such as diagnostic failures and protracted treatment regimens continue to impede progress. This work aims to steer future research endeavors toward groundbreaking TB vaccines and therapeutic agents, providing crucial insights for enhancing TB prevention and treatment strategies.
Collapse
Affiliation(s)
- Yajing An
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, 17#Heishanhu Road, Haidian District, Beijing, 100091, China
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ruizi Ni
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Li Zhuang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ling Yang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Zhaoyang Ye
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Linsheng Li
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, 33014, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories PLC, Tampere, Finland
| | - Ashok Aspatwar
- Faculty of Medicine and Health Technology, Tampere University, 33014, Tampere, Finland.
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, 17#Heishanhu Road, Haidian District, Beijing, 100091, China.
| |
Collapse
|
2
|
Faisal S, Vats D, Panda S, Sharma V, Luthra K, Mohan A, Kulkarni S, Gupta PK, Singh A. Synergistic role of Mycobacterium indicus pranii and human beta Defensin-2 as adjunctive therapy against Mycobacterium tuberculosis. Tuberculosis (Edinb) 2024; 149:102571. [PMID: 39442483 DOI: 10.1016/j.tube.2024.102571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Host-directed therapies (HDT) via modulation of specific host responses like inflammation can limit mycobacterial infection. HDTs could be included in current TB therapy as an adjunct to increase bacterial clearance and limit tissue damage to control spread. Individually, Mycobacterium indicus pranii (MIP) and human beta defensin-2 (hBD-2) are promising therapies for tuberculosis (TB). They can directly target the TB bacilli and enhance cell-mediated immune responses, which is limiting with conventional drugs. Therefore, our study investigated the combined application of MIP and hBD-2 to evaluate their efficacy in clearing infections caused by Mycobacterium smegmatis (M.smeg) and Mycobacterium tuberculosis (M.tb) (both avirulent; H37Ra and virulent strain; H37Rv) in THP-1 cells and human monocyte-derived macrophages (MDMs). A strong pro-inflammatory response was observed against the combination of MIP and hBD-2 which also correlated with a significant reduction in the bacterial load. This combination further showed protection against M.tb by enhancing pyroptosis in the infected cells. The study suggests the combined use of these potent immunomodulators, which could be employed as an effective mode of therapy as adjuvants against mycobacterial infections after validation in a suitable animal model.
Collapse
Affiliation(s)
- Shah Faisal
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Deepak Vats
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sudhasini Panda
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Vidushi Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Anant Mohan
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Savita Kulkarni
- Molecular Immunology and Tuberculosis Section, RMC, BARC, Mumbai, 400012, India
| | - Pramod Kumar Gupta
- Molecular Immunology and Tuberculosis Section, RMC, BARC, Mumbai, 400012, India.
| | - Archana Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
3
|
Stefan K, Gordon R, Rolig A, Honkala A, Tailor D, Davis LE, Modi RI, Joshipura M, Khamar B, Malhotra SV. Mycobacterium w - a promising immunotherapeutic intervention for diseases. Front Immunol 2024; 15:1450118. [PMID: 39534596 PMCID: PMC11554463 DOI: 10.3389/fimmu.2024.1450118] [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: 06/16/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
Immunomodulating agents interact with the immune system and alter the outcome of specific immune processes. As our understanding of the immune system continues to evolve, there is a growing effort to identify agents with immunomodulating applications to use therapeutically to treat various diseases. Mycobacterium w (Mw), a heat-killed mycobacterium, is an atypical mycobacterial species that possesses strong immunomodulatory properties. Mw was initially evaluated as an immune-therapeutic against leprosy, but since then Mw has generated a lot of interest and been studied for therapeutic applications across a host of diseases, such as pulmonary tuberculosis, tuberculous pericarditis, sepsis, lung cancer, and more. This article summarizes a large body of work published in the past five decades, describing various aspects of Mw and its potential for further therapeutic development.
Collapse
Affiliation(s)
- Kirsten Stefan
- Department of Cell, Development & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Ryan Gordon
- Department of Cell, Development & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Annah Rolig
- Department of Cell, Development & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Alexander Honkala
- Department of Cell, Development & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Dhanir Tailor
- Department of Cell, Development & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Lara E. Davis
- Division of Hematology/Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Rajiv I. Modi
- Research & Development Center, Cadila Pharmaceuticals Ltd, Dholka, Gujarat, India
| | - Manjul Joshipura
- Research & Development Center, Cadila Pharmaceuticals Ltd, Dholka, Gujarat, India
| | - Bakulesh Khamar
- Research & Development Center, Cadila Pharmaceuticals Ltd, Dholka, Gujarat, India
| | - Sanjay V. Malhotra
- Department of Cell, Development & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
4
|
Liu J, Chen Y, Laurent I, Yang P, Xiao X, Li X. Gestational diabetes exacerbates intrauterine microbial exposure induced intestinal microbiota change in offspring contributing to increased immune response. Nutr Diabetes 2024; 14:87. [PMID: 39424815 PMCID: PMC11489853 DOI: 10.1038/s41387-024-00346-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 09/20/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND maternal health during pregnancy can affect the intestinal microbial community of offspring, but currently the impact of intrauterine environmental changes resulting from gestational diabetes mellitus (GDM) on the microbiota of offspring as well as its interaction with the immune system remains unclear. AIMS to explore the impact of intrauterine microbial exposure during pregnancy of gestational diabetes mellitus on the development of neonate's intestinal microbiota and activation of immune responses. METHODS Levels of lipopolysaccharides in cord blood from GDM and expression of microbial recognition-related proteins in the placenta were measured. To evaluate embryonic intestinal colonization, pregnant mice with GDM were administered with labeled Escherichia coli or Lactobacillus. The intestinal colonization of pups was analyzed through 16S rRNA gene sequencing and labeled microbial culture. Additionally, memory T lymphocyte and dendritic cell co-culture experiments were conducted to elucidate the immune memory of intestinal microbes during the embryonic stages. RESULT Gestational diabetes mellitus led to elevated umbilical cord blood LPS level and increased GFP labeled Escherichia coli in the offspring's intestine after gestational microbial exposure. The mouse model of GDM exhibited increased immune markers including TLR4, TLR5, IL-22 and IL-23 in the placenta and a recall response from memory T cells in offspring's intestines, with similar observations found in human experiments. Furthermore, reduced intestinal microbiome diversity and an increased ratio of Firmicutes/Bacteroidetes was found in GDM progeny, with the stability of bacterial colonization been interfered. CONCLUSIONS Our investigation has revealed a noteworthy correlation between gestational diabetes and intrauterine microbial exposure, as well as alterations in the neonatal microbiota and activation of immune responses. These findings highlight the gestational diabetes's role on offspring's gut microbiota and immune system interactions with early-life pathogen exposure.
Collapse
Affiliation(s)
- Juncheng Liu
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yan Chen
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Endocrinology and Nephrology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Irakoze Laurent
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Yang
- Yongchuan Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Xiaoqiu Xiao
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Xinyu Li
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Department of Pharmacy, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
5
|
Dogra S, Jain S, Sharma A, Chhabra S, Narang T. Mycobacterium Indicus Pranii (MIP) Vaccine: Pharmacology, Indication, Dosing Schedules, Administration, and Side Effects in Clinical Practice. Indian Dermatol Online J 2023; 14:753-761. [PMID: 38099011 PMCID: PMC10718117 DOI: 10.4103/idoj.idoj_360_23] [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: 05/09/2023] [Revised: 08/07/2023] [Accepted: 08/19/2023] [Indexed: 12/17/2023] Open
Abstract
Mycobacterium indicus pranii (MIP), previously called Mw vaccine, is a one-of-a-kind immunomodulatory vaccine. It was indigenously developed in India for use in leprosy. MIP is heat-killed Mycobacterium w, which is a non-pathogenic atypical mycobacterium belonging to Class IV of Runyon classification. It shares epitopes with Mycobacterium leprae and Mycobacterium tuberculosis, which forms the rationale behind its use in leprosy and tuberculosis. MIP activates both innate and acquired immunity. It induces a Th1 and Th17 immune response along with downregulation of Th2 pathway and activates macrophages and dendritic cells. MIP vaccine is safe with adverse effects such as local site erythema, swelling, and rarely fever and other systemic reactions. Apart from leprosy, MIP has been used in dermatological diseases such as warts and psoriasis. Clinical trials have evaluated the efficacy of MIP in a plenitude of non-dermatological conditions such as category II tuberculosis, Gram-negative sepsis, non-small cell lung cancer, human immunodeficiency virus (HIV), muscle-invasive bladder cancer, and very recently, coronavirus 2019 (COVID-19). In vitro and animal studies have also demonstrated its utility in leishmaniasis, melanoma, and as a vaccine for the prevention of pregnancy. The PubMed database was searched using "Mycobacterium indicus pranii, MIP, Mycobacterium w" as the keyword in title. This comprehensive review provides useful information for healthcare professionals about immunotherapeutic potential of MIP vaccine, its composition, dosing schedule, administration, and side effects besides its efficacy in various indications other than leprosy.
Collapse
Affiliation(s)
- Sunil Dogra
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sejal Jain
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ayush Sharma
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Seema Chhabra
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Tarun Narang
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
6
|
Chakraborty A, Roy G, Swami B, Bhaskar S. Tumor targeted delivery of mycobacterial adjuvant encapsulated chitosan nanoparticles showed potential anti-cancer activity and immune cell activation in tumor microenvironment. Int Immunopharmacol 2023; 114:109463. [PMID: 36462337 DOI: 10.1016/j.intimp.2022.109463] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/01/2022] [Accepted: 11/12/2022] [Indexed: 12/03/2022]
Abstract
Targeting immunotherapeutics inside the tumor microenvironment (TME) with intact biological activity remains a pressing issue. Mycobacterium indicus pranii (MIP), an approved adjuvant therapy for leprosy has exhibited promising results in clinical trials of lung (NSCLC) and bladder cancer. Whole MIP as well as its cell wall fraction have shown tumor growth suppression and enhanced survival in mice model of melanoma, when administered peritumorally. Clinically, peritumoral delivery remains a procedural limitation. In this study, a tumor targeted delivery system was designed, where chitosan nanoparticles loaded with MIP adjuvants, when administered intravenously showed preferential accumulation within the TME, exploiting the principle of enhanced permeability and retention effect. Bio-distribution studies revealed their highest concentration inside the tumor after 6 h of administration. Interestingly, MIP adjuvant nano-formulations significantly reduced the tumor volume in the treated groups and increased the frequency of activated immune cells inside the TME. For chemoimmunotherapeutics studies, MIP nano-formulation was combined with standard dosage regimen of Paclitaxel. Combined therapy exhibited a further reduction in tumor volume relative to either of the MIP nano formulations. From this study a three-pronged strategy emerged as the underlying mechanism; chitosan and Paclitaxel have shown direct role in tumor cell death and the MIP nano-formulation activates the tumor residing immune cells which ultimately leads to the reduced tumor growth.
Collapse
Affiliation(s)
- Anush Chakraborty
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Gargi Roy
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Bharati Swami
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Sangeeta Bhaskar
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
7
|
Roy G, Chakraborty A, Swami B, Pal L, Ahuja C, Basak S, Bhaskar S. Type 1 interferon mediated signaling is indispensable for eliciting anti-tumor responses by Mycobacterium indicus pranii. Front Immunol 2023; 14:1104711. [PMID: 37122749 PMCID: PMC10140407 DOI: 10.3389/fimmu.2023.1104711] [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: 11/21/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction The evolving tumor secretes various immunosuppressive factors that reprogram the tumor microenvironment (TME) to become immunologically cold. Consequently, various immunosuppressive cells like Tregs are recruited into the TME which in turn subverts the anti-tumor response of dendritic cells and T cells.Tumor immunotherapy is a popular means to rejuvenate the immunologically cold TME into hot. Mycobacterium indicus pranii (MIP) has shown strong immunomodulatory activity in different animal and human tumor models and has been approved for treatment of lung cancer (NSCLC) patients as an adjunct therapy. Previously, MIP has shown TLR2/9 mediated activation of antigen presenting cells/Th1 cells and their enhanced infiltration in mouse melanoma but the underlying mechanism by which it is modulating these immune cells is not yet known. Results This study reports for the first time that MIP immunotherapy involves type 1 interferon (IFN) signaling as one of the major signaling pathways to mediate the antitumor responses. Further, it was observed that MIP therapy significantly influenced frequency and activation of different subsets of T cells like regulatory T cells (Tregs) and CD8+ T cells in the TME. It reduces the migration of Tregs into the TME by suppressing the expression of CCL22, a Treg recruiting chemokine on DCs and this process is dependent on type 1 IFN. Simultaneously, in a type 1 IFN dependent pathway, it enhances the activation and effector function of the immunosuppressive tumor resident DCs which in turn effectively induce the proliferation and effector function of the CD8+ T cells. Conclusion This study also provides evidence that MIP induced pro-inflammatory responses including induction of effector function of conventional dendritic cells and CD8+ T cells along with reduction of intratumoral Treg frequency are essentially mediated in a type 1 IFN-dependent pathway.
Collapse
Affiliation(s)
- Gargi Roy
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Anush Chakraborty
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Bharati Swami
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Lalit Pal
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Charvi Ahuja
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Soumen Basak
- Systems Immunology Lab, National Institute of Immunology, New Delhi, India
| | - Sangeeta Bhaskar
- Product Development Cell, National Institute of Immunology, New Delhi, India
- *Correspondence: Sangeeta Bhaskar,
| |
Collapse
|
8
|
Microbial-Derived Toll-like Receptor Agonism in Cancer Treatment and Progression. Cancers (Basel) 2022; 14:cancers14122923. [PMID: 35740589 PMCID: PMC9221178 DOI: 10.3390/cancers14122923] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/02/2022] [Accepted: 06/13/2022] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Toll like receptors (TLRs) are a group of transmembrane receptors belonging to the class of pattern recognition receptors (PRR), which are involved in recognition of pathogen associated molecular patterns (PAMPs), inducing immune response. During the past decade, a number of preclinical and clinical breakthroughs in the field of TLR agonists has immerged in cancer research and some of these agents have performed exceptionally well in clinical trials. Based on evidence from scientific studies, we draw attention to several microbial based TLR agonists and discuss their relevance in various cancer and explore various microbial based TLR agonists for developing effective immunotherapeutic strategies against cancer. Abstract Toll-like receptors (TLRs) are typical transmembrane proteins, which are essential pattern recognition receptors in mediating the effects of innate immunity. TLRs recognize structurally conserved molecules derived from microbes and damage-associated molecular pattern molecules that play an important role in inflammation. Since the first discovery of the Toll receptor by the team of J. Hoffmann in 1996, in Drosophila melanogaster, numerous TLRs have been identified across a wide range of invertebrate and vertebrate species. TLR stimulation leads to NF-κB activation and the subsequent production of pro-inflammatory cytokines and chemokines, growth factors and anti-apoptotic proteins. The expression of TLRs has also been observed in many tumors, and their stimulation results in tumor progression or regression, depending on the TLR and tumor type. The anti-tumoral effects can result from the activation of anti-tumoral immune responses and/or the direct induction of tumor cell death. The pro-tumoral effects may be due to inducing tumor cell survival and proliferation or by acting on suppressive or inflammatory immune cells in the tumor microenvironment. The aim of this review is to draw attention to the effects of TLR stimulation in cancer, the activation of various TLRs by microbes in different types of tumors, and, finally, the role of TLRs in anti-cancer immunity and tumor rejection.
Collapse
|
9
|
Jaiswal SR, Arunachalam J, Saifullah A, Lakhchaura R, Tailor D, Mehta A, Bhagawati G, Aiyer H, Khamar B, Malhotra SV, Chakrabarti S. Impact of an Immune Modulator Mycobacterium-w on Adaptive Natural Killer Cells and Protection Against COVID-19. Front Immunol 2022; 13:887230. [PMID: 35603154 PMCID: PMC9115578 DOI: 10.3389/fimmu.2022.887230] [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: 03/01/2022] [Accepted: 04/04/2022] [Indexed: 11/23/2022] Open
Abstract
The kinetics of NKG2C+ adaptive natural killer (ANK) cells and NKG2A+inhibitory NK (iNK) cells with respect to the incidence of SARS-CoV-2 infection were studied for 6 months in a cohort of healthcare workers following the administration of the heat-killed Mycobacterium w (Mw group) in comparison to a control group. In both groups, corona virus disease 2019 (COVID-19) correlated with lower NKG2C+ANK cells at baseline. There was a significant upregulation of NKG2C expression and IFN-γ release in the Mw group (p=0.0009), particularly in those with a lower baseline NKG2C expression, along with the downregulation of iNK cells (p<0.0001). This translated to a significant reduction in the incidence and severity of COVID-19 in the Mw group (incidence risk ratio-0.15, p=0.0004). RNA-seq analysis at 6 months showed an upregulation of the ANK pathway genes and an enhanced ANK-mediated antibody-dependent cellular cytotoxicity (ADCC) signature. Thus, Mw was observed to have a salutary impact on the ANK cell profile and a long-term upregulation of ANK-ADCC pathways, which could have provided protection against COVID-19 in a non-immune high-risk population.
Collapse
Affiliation(s)
- Sarita Rani Jaiswal
- Cellular Therapy and Immunology, Manashi Chakrabarti Foundation, New Delhi, India
- Department of Blood and Marrow Transplantation, Dharamshila Narayana Super-Speciality Hospital, New Delhi, India
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Jaganath Arunachalam
- Cellular Therapy and Immunology, Manashi Chakrabarti Foundation, New Delhi, India
| | - Ashraf Saifullah
- Department of Blood and Marrow Transplantation, Dharamshila Narayana Super-Speciality Hospital, New Delhi, India
| | - Rohit Lakhchaura
- Department of Blood and Marrow Transplantation, Dharamshila Narayana Super-Speciality Hospital, New Delhi, India
| | - Dhanir Tailor
- Department of Cell, Development & Cancer Biology and Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Anupama Mehta
- Department of Blood and Marrow Transplantation, Dharamshila Narayana Super-Speciality Hospital, New Delhi, India
| | - Gitali Bhagawati
- Department of Pathology and Microbiology, Dharamshila Narayana Super-speciality Hospital, New Delhi, India
| | - Hemamalini Aiyer
- Department of Pathology and Microbiology, Dharamshila Narayana Super-speciality Hospital, New Delhi, India
| | - Bakulesh Khamar
- Research & Development, Cadila Pharmaceuticals Ltd, Ahmedabad, India
| | - Sanjay V. Malhotra
- Department of Cell, Development & Cancer Biology and Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Suparno Chakrabarti
- Cellular Therapy and Immunology, Manashi Chakrabarti Foundation, New Delhi, India
- Department of Blood and Marrow Transplantation, Dharamshila Narayana Super-Speciality Hospital, New Delhi, India
| |
Collapse
|
10
|
Abstract
COVID-19 has become a major pandemic in recent times. The exact pathophysiology and understanding of cytokine storm and immunomodulation are evolving. Various cytokines have been implicated in the pathophysiology of COVID-19. Immunosuppressant immunomodulators like steroids, canakinumab, anakinra, tocilizumab, sarilumab, baricitinib, ruxolitinib, bevacizumab, and itolizumab have been tried. Immunostimulant immunomodulators like interferons (IFNs) and Mycobacterium w (Mw) have also been repurposed. Considering the role of multiple cytokines implicated in COVID-19, molecules working on the majority of the targets, may hold a promising future prospect
Collapse
Affiliation(s)
- Pradeep Rangappa
- Department of Critical Care Medicine, Columbia Asia, Bengaluru, Karnataka, India
| |
Collapse
|
11
|
Chen HM, Sun L, Pan PY, Wang LH, Chen SH. Nutrient supplements from selected botanicals mediated immune modulation of the tumor microenvironment and antitumor mechanism. Cancer Immunol Immunother 2021; 70:3435-3449. [PMID: 33877384 DOI: 10.1007/s00262-021-02927-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/23/2021] [Indexed: 11/29/2022]
Abstract
Specific extracts of selected vegetables (SV) have been shown to benefit the survival of stage IIIb/IV non-small cell lung cancer patients in phase I/II studies and is currently in a phase III trial. However, the underlying mechanism of SV-mediated antitumor immune responses has not been elucidated. Our results indicate that SV modulated the NK and adoptive T cell immune responses in antitumor efficacy. Furthermore, antitumor effects of SV were also mediated by innate myeloid cell function, which requires both TLR and β-glucan signaling in a MyD88/TRIF and Dectin-1-dependent manner, respectively. Additionally, SV treatment reduced granulocytic myeloid-derived suppressor cell (MDSC) infiltration into the tumor and limited monocytic MDSC toward the M2-like functional phenotype. Importantly, SV treatment enhanced antigen-specific immune responses by augmenting the activation of antigen-specific TH1/TH17 cells in secondary lymphoid organs and proliferative response, as well as by reducing the Treg population in the tumor microenvironment, which was driven by SV-primed activated M-MDSC. Our results support the idea that SV can subvert immune-tolerance state in the tumor microenvironment and inhibit tumor growth. The present study suggests that features, such as easy accessibility, favorable clinical efficacy, no detectable side effects and satisfactory safety make SV a feasible, appealing and convincing adjuvant therapy for the treatment of cancer patients and prevent tumor recurrence and/or metastases.
Collapse
Affiliation(s)
- Hui-Ming Chen
- Department of Oncological Sciences, Icahn School of Medicine At Mount Sinai, New York, 10029, USA
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Cancer Center, Houston Methodist Research Institute, Houston, 77030, USA
| | - Linus Sun
- Department of Ophthalmology, Columbia University, New York, 10027, USA
| | - Ping-Ying Pan
- Department of Oncological Sciences, Icahn School of Medicine At Mount Sinai, New York, 10029, USA
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Cancer Center, Houston Methodist Research Institute, Houston, 77030, USA
| | - Lu-Hai Wang
- Institute of Integrated Medicine and Chinese Medical Research Center, China Medical University, Taichung, 40402, Taiwan, ROC.
| | - Shu-Hsia Chen
- Department of Oncological Sciences, Icahn School of Medicine At Mount Sinai, New York, 10029, USA.
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Cancer Center, Houston Methodist Research Institute, Houston, 77030, USA.
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, USA.
| |
Collapse
|
12
|
Zheng C, Chen J, Chu F, Zhu J, Jin T. Inflammatory Role of TLR-MyD88 Signaling in Multiple Sclerosis. Front Mol Neurosci 2020; 12:314. [PMID: 31998072 PMCID: PMC6965019 DOI: 10.3389/fnmol.2019.00314] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a neuro-autoimmune and neurodegenerative disorder leading to chronic inflammation, demyelination, axonal, and neuronal loss in the central nervous system (CNS). Despite intense research efforts, the pathogenesis of MS still remains unclear. Toll-like receptors (TLRs) are a family of type I transmembrane receptors that play a crucial role in the innate immune response. Myeloid differentiation factor 88 (MyD88) is the adaptor of major TLRs. It has been widely considered that the TLR-MyD88 signaling pathway plays an important role in the occurrence and development of autoimmune disease. Data have revealed that the TLR-MyD88 signaling may be involved in the pathogenesis of MS and experimental autoimmune encephalomyelitis (EAE), an animal model for MS, by regulating the antigen presentation of dendritic cells, the integrity of blood-brain barrier (BBB), and the activation of T cells and B cells. Here, we summarize the role of TLRs and MyD88 in MS and discuss the possible therapies that are based on these molecules.
Collapse
Affiliation(s)
- Chao Zheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Fengna Chu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Kumar P, Das G, Bhaskar S. Mycobacterium indicus pranii therapy induces tumor regression in MyD88- and TLR2-dependent manner. BMC Res Notes 2019; 12:648. [PMID: 31590685 PMCID: PMC6781299 DOI: 10.1186/s13104-019-4679-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 09/28/2019] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVES Mycobacterium indicus pranii (MIP) is an atypical mycobacterium species with potent antitumor efficacy. Macrophages and dendritic cells (DCs) are antigen-presenting cells, playing key roles in the activation of antitumor immunity. We have previously shown the potent activation of macrophages and DCs by MIP, which is mediated by MyD88-TLR2 signaling axis. In the present study, we further examined the role of MyD88 and TLR2 in MIP-mediated tumor regression. RESULTS Wild-type and MyD88-/- mice were implanted with B16F10 tumor cells, treated with MIP or phosphate-buffered saline (PBS) and monitored for tumor growth. As expected, MIP therapy led to significant tumor regression in wild-type mice. However, antitumor efficacy of MIP was lost in MyD88-/- animals. Both PBS-treated (control) and MIP-treated MyD88-/- mice developed tumors with comparable volume. Since MyD88 relays TLR engagement signals, we analyzed the antitumor efficacy of MIP in TLR2-/- and TLR4-/- mice. It was observed that MIP therapy reduced tumor burden in wild-type and TLR4-/- mice but not in TLR2-/- mice. Tumor volume in MIP-treated TLR2-/- mice were comparable with those in PBS-treated wild-type animals. These results implicated the MyD88-TLR2 signaling axis in the antitumor efficacy of MIP.
Collapse
Affiliation(s)
- Pawan Kumar
- PDC-I, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India.,Dept. of Preventive Oncology, Dr. B. R. Ambedkar Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Gobardhan Das
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sangeeta Bhaskar
- PDC-I, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India.
| |
Collapse
|
14
|
Kumar P, Bhaskar S. Myeloid differentiation primary response protein 88 (MyD88)-deficient dendritic cells exhibit a skewed cytokine response to BCG. BMC Res Notes 2019; 12:52. [PMID: 30674337 PMCID: PMC6343232 DOI: 10.1186/s13104-019-4086-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/14/2019] [Indexed: 11/29/2022] Open
Abstract
Objective Macrophages and dendritic cells (DCs) play key role in the recognition of mycobacterial infection and mounting of antimycobacterial immunity. In case of macrophages, recognition of BCG and other mycobacteria has been attributed predominantly to MyD88-dependent singling. Interestingly, in previous study with bone marrow-derived DCs, we have shown that BCG promotes the survival of wild-type and MyD88−/− cells to the comparable levels. In the present study, we further examined MyD88−/− DC’s response to BCG. Results Bone marrow-derived DCs from wild-type and MyD88−/− mice were stimulated with BCG and analyzed for cytokine secretion. As expected, BCG-stimulated wild-type DCs produced significant amount of TNF-α and IL-12p40 in response to BCG. Interestingly, BCG-stimulated MyD88−/− DCs were also found to secret significantly higher levels of TNF-α and IL-12p40, compared with unstimulated DCs. We further observed that wild-type DCs produced significant level of immunosuppressive cytokine IL-10 in response to BCG, whereas MyD88−/− DCs secreted very low amount of IL-10 when stimulated with BCG. These findings demonstrated that MyD88−/− DCs exhibit a skewed cytokine response to BCG.
Collapse
Affiliation(s)
- Pawan Kumar
- Dept. of Preventive Oncology, Dr. B.R Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.,PDC-I, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Sangeeta Bhaskar
- PDC-I, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India.
| |
Collapse
|
15
|
Kumar P, John V, Gupta A, Bhaskar S. Enhanced survival of BCG-stimulated dendritic cells: involvement of anti-apoptotic proteins and NF-κB. Biol Open 2018; 7:bio.032045. [PMID: 29848490 PMCID: PMC6031337 DOI: 10.1242/bio.032045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BCG (Bacillus Calmette-Guérin) is the only available vaccine against TB and is also used for the treatment of superficial bladder cancer. BCG-mediated protection against TB and bladder cancer has been shown to rely on its ability to induce superior CD4+ and CD8+ T cell responses. As the magnitude of T cell responses is defined by dendritic cell (DC) lifespan, we examined the effect of BCG on DC survival and its underlying mechanisms. It was observed that BCG stimulation enhanced DC survival and prolonged DC lifespan in a dose-dependent manner. Live BCG led to a higher DC survival compared with heat-killed BCG. FITC-Annexin V staining showed that BCG promoted DC survival by inhibiting apoptosis. Consistently, higher expressions of anti-apoptotic proteins Bcl-2 and Bcl-xL were observed in BCG-stimulated DCs. Pharmacological inhibition of Bcl-2 and Bcl-xL drastically reduced the DC survival efficacy of BCG. Comparable survival of BCG-stimulated wild-type and MyD88−/− DCs suggested that MyD88 signaling is dispensable for BCG-induced DC survival. NF-κB is one of the key regulators of innate immune responses. We observed that pharmacological inhibition of NF-κB abrogated BCG-mediated increase in DC survival and expression of anti-apoptotic proteins. These findings provide a novel insight into the effect of BCG on DC physiology. Summary: BCG enhanced the survival of dendritic cells (DCs) and prolonged their lifespan. BCG promoted DC survival by up-regulating Bcl-2 and Bcl-xL. Increased survival of BCG-stimulated DCs was dependent on NF-κB, but was independent of MyD88 signaling.
Collapse
Affiliation(s)
- Pawan Kumar
- Product Development Cell-I, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Vini John
- Product Development Cell-I, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Ananya Gupta
- Product Development Cell-I, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Sangeeta Bhaskar
- Product Development Cell-I, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| |
Collapse
|
16
|
Garg H, Gupta JC, Talwar GP, Dubey S. Immunotherapy approach with recombinant survivin adjuvanted with alum and MIP suppresses tumor growth in murine model of breast cancer. Prep Biochem Biotechnol 2018; 48:264-269. [PMID: 29355462 DOI: 10.1080/10826068.2018.1425710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Survivin has received attention as a potential target for cancer immunotherapy because of its crucial role in oncogenesis. We undertook this study to evaluate the immunotherapeutic potential of combination of recombinant survivin along with adjuvant alum and immune modulator Mycobacterium indicus pranii (MIP). In vivo efficacy of the combination was studied in an invasive murine breast cancer model. Recombinant survivin protein was purified from Escherichia coli based expression system and characterized by western blotting. Purified survivin protein was combined with alum and MIP and was used for immunization of Balb/c mice. Antigen-primed animals were then challenged with syngeneic mammary tumor cells known as 4T-1. Balb/c mice spontaneously develop tumor when inoculated with 4T-1 cells. Antigen and adjuvant combination was immunogenic and significantly suppressed tumor growth in mice immunized with combination of recombinant survivin (10 µg), alum, and MIP. This is the first report that describes a combination immunotherapy approach using recombinant survivin, alum, and MIP in highly metastatic murine breast cancer model and holds promise for development of new biotherapeutics for cancer.
Collapse
Affiliation(s)
- Himani Garg
- a Amity Institute of Virology and Immunology , Amity University, Uttar Pradesh , Noida , India.,b Talwar Research Foundation, Neb Sarai , New Delhi , India
| | | | - G P Talwar
- b Talwar Research Foundation, Neb Sarai , New Delhi , India
| | - Shweta Dubey
- a Amity Institute of Virology and Immunology , Amity University, Uttar Pradesh , Noida , India
| |
Collapse
|
17
|
Garg H, Suri P, Gupta JC, Talwar GP, Dubey S. Survivin: a unique target for tumor therapy. Cancer Cell Int 2016; 16:49. [PMID: 27340370 PMCID: PMC4917988 DOI: 10.1186/s12935-016-0326-1] [Citation(s) in RCA: 321] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/10/2016] [Indexed: 12/13/2022] Open
Abstract
Survivin is the smallest member of the Inhibitor of apoptosis (IAP) family of proteins, involved in inhibition of apoptosis and regulation of cell cycle. These functional attributes make Survivin a unique protein exhibiting divergent functions i.e. regulating cell proliferation and cell death. Expression pattern of Survivin is also distinctive; it is prominently expressed during embryonal development, absent in most normal, terminally differentiated tissues but upregulated in a variety of human cancers. Expression of Survivin in tumours correlates with not only inhibition of apoptosis and a decreased rate of cell death, but also resistance to chemotherapy and aggressiveness of tumours. Therefore, Survivin is an important target for cancer vaccines and therapeutics. Survivin has also been found to be prominently expressed on both human and embryonic stem cells and many somatic stem cell types indicating its yet unexplored role in stem cell generation and maintenance. Overall, Survivin emerges as a molecule with much wider role in cellular homeostasis. This review will discuss various aspects of Survivin biology and its role in regulation of apoptosis, cell division, chemo-resistance and tumour progression. Various molecular and immunotherapeutic approaches targeting Survivin will also be discussed.
Collapse
Affiliation(s)
- Himani Garg
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, J-3 Block, Room No: LG21, Sector 125, Noida, Uttar Pradesh 201303 India
| | - Prerna Suri
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Sector 125, Noida, India
| | - Jagdish C Gupta
- Talwar Research Foundation, E-8 Neb Valley, Neb Sarai, New Delhi, 110 068 India
| | - G P Talwar
- Talwar Research Foundation, E-8 Neb Valley, Neb Sarai, New Delhi, 110 068 India
| | - Shweta Dubey
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, J-3 Block, Room No: LG21, Sector 125, Noida, Uttar Pradesh 201303 India
| |
Collapse
|
18
|
Lee GJ, Lee HM, Kim TS, Kim JK, Sohn KM, Jo EK. Mycobacterium fortuitum induces A20 expression that impairs macrophage inflammatory responses. Pathog Dis 2016; 74:ftw015. [PMID: 26940588 DOI: 10.1093/femspd/ftw015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2016] [Indexed: 12/30/2022] Open
Abstract
Mycobacterium fortuitum is a rapidly growing mycobacterium that has been regarded as an etiological agent of a variety of human infections. However, little is known about the host inflammatory responses and the molecular mechanisms by which MF-induced inflammation is regulated in macrophages. In this study, we report that MF infection leads to the induction of an anti-inflammatory molecule, A20 (also known as TNFAIP3), which is essential for the regulation of MF-induced inflammatory responses in murine bone marrow-derived macrophages (BMDMs). MF triggered the expression of tumor necrosis factor-α and interleukin-6 in BMDMs through signaling of the Toll-like receptor 2 (TLR2)-myeloid differentiation primary response gene 88. Additionally, MF rapidly induced the expression of A20, which inhibited proinflammatory cytokine expression and nuclear factor (NF)-κB reporter gene activities in BMDMs. Notably, MF-induced activation of NF-κB signaling was required for A20 expression and proinflammatory responses in BMDMs. Furthermore, the rough morphotype of the MF clinical strain induced a higher level of proinflammatory signaling activation, but less A20 induction in BMDMs, compared to the smooth morphotype. Taken together, these results suggest that MF-induced activation of host proinflammatory responses is negatively regulated through TLR2-dependent A20 expression.
Collapse
Affiliation(s)
- Gippeum Joy Lee
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea
| | - Hye-Mi Lee
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea
| | - Tae Sung Kim
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea
| | - Kyung Mok Sohn
- Division of Infectious Diseases, Chungnam National University Hospital, Daejeon 35015, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea
| |
Collapse
|
19
|
|
20
|
Kumar P, Bhaskar S. Analysis of T Cell Proliferating and Polarizing Potential of Murine Dendritic Cells in Allogeneic-mixed Leukocyte Reaction. Bio Protoc 2016. [DOI: 10.21769/bioprotoc.1750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|