1
|
Han X, Wang Y, Zhang K, Na T, Wu T, Hao X, Jin Y, Wang Y, Wang H, Meng S. Dosage and organic acid residue of myelin oligodendrocyte glycoprotein 35-55 peptide influences immunopathology and development of Bacillus Calmette-Guérin induced experimental autoimmune encephalomyelitis. Exp Anim 2025; 74:16-30. [PMID: 38987201 PMCID: PMC11742474 DOI: 10.1538/expanim.24-0012] [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: 02/07/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) serves as a model for studying multiple sclerosis, with immunization strategies utilizing myelin oligodendrocyte glycoprotein (MOG)35-55 peptide, emulsified in adjuvant enriched with Mycobacterium tuberculosis (Mtb). This study examined the effects of Bacillus Calmette-Guérin (BCG) as an adjuvant, alongside the impact of MOG35-55 peptide doses and their residual counter ions on EAE development. We found that BCG can be effectively used to induce EAE with similar incidence and severity as heat-killed H37Ra, contingent upon the appropriate MOG35-55 peptide dose. Different immunization doses of MOG35-55 peptide significantly affect EAE development, with higher doses leading to a paradoxical reduction in disease activity, probably due to peripheral tolerance mechanisms. Furthermore, doses of MOG35-55 peptides with acetate showed a more pronounced effect on disease development compared to those containing trifluoroacetic acid (TFA), suggesting the potential influence of residual counter ions on EAE activity. We highlighted the feasibility of applying BCG to the establishment of EAE for the first time. Our findings emphasized the importance of MOG35-55 peptide dosage and composition in modulating EAE development, offering insights into the mechanisms of autoimmunity and tolerance. This could have implications for autoimmune disease research and the design of therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoyan Han
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Ying Wang
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Kehua Zhang
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Tao Na
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Tingting Wu
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Xiaofang Hao
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Yuxuan Jin
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Yuchun Wang
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Haohan Wang
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Shufang Meng
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| |
Collapse
|
2
|
Yedke NG, Soni D, Kumar P. Effect of Bacille-Calmette-Guerin vaccine against rotenone-induced Parkinson's disease: Role of neuroinflammation and neurotransmitters. Fundam Clin Pharmacol 2024; 38:538-549. [PMID: 38041521 DOI: 10.1111/fcp.12968] [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: 01/20/2023] [Revised: 10/22/2023] [Accepted: 10/30/2023] [Indexed: 12/03/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is an extrapyramidal movement disorder associated with a hypokinetic condition generated by impairment in dopaminergic neuronal viability in the nigrostriatal region of the brain. Current medications can only provide symptomatic management; to date, no permanent cure is available. To compensate for this lacuna, researchers are gaining interest in antigen-based therapy, and Bacille-Calmette-Guerin (BCG) is one of the vaccines with a high safety margin that acts by stimulating immunoreactive T-cells in the CNS and reducing expression of pro-inflammatory cytokines including interleukin (IL)-1β and tumor necrotic factor (TNF-α) to produce neuroprotection. A previous study reported that BCG exerts a neuroprotective effect against several neurodegenerative disorders, such as Alzheimer's disease. OBJECTIVE The objective of this study is to explore the neuroprotective effect of the BCG vaccine against the rotenone model of PD. METHODS Rotenone (1.5 mg/kg, s.c) for 28 days, and BCG vaccine (2 × 107 cfu, i.p) single dose was injected to rats, and behavioral assessments were performed on the 21st and 28th day. On the 29th day, rats were sacrificed, and brains were isolated for biochemical and neurochemical estimation. RESULTS BCG vaccine significantly restored rotenone-induced motor deficits (open field test, narrow beam walk, and rotarod), biochemical levels (GSH, SOD, catalase, MDA, and nitrite), neurotransmitters (dopamine, 5-hydroxy tryptamine, norepinephrine, 3,4-dihydroxyphenylacetic acid, hemovanillic acid, and 5-hydroxy indoleacetic acid), and levels of inflammatory cytokines (IL-1β and TNF-α) in the striatum. It also prevents histopathological changes by reducing eosinophilic lesions in the striatum. CONCLUSION From the results, we conclude that BCG vaccine showed neuroprotection through antioxidant and anti-inflammatory effect. Thus, in the future, it can be used as a neuroprotective agent for other neurological disorders, including PD.
Collapse
Affiliation(s)
- Narhari Gangaram Yedke
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, India
| | - Divya Soni
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|
3
|
Liu X, Liu M, Zhao M, Li P, Gao C, Fan X, Cai G, Lu Q, Chen X. Fecal microbiota transplantation for the management of autoimmune diseases: Potential mechanisms and challenges. J Autoimmun 2023; 141:103109. [PMID: 37690971 DOI: 10.1016/j.jaut.2023.103109] [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: 01/17/2023] [Revised: 08/17/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023]
Abstract
Autoimmune diseases (AIDs) are a series of immune-mediated lethal diseases featured by over-activated immune cells attacking healthy self-tissues and organs due to the loss of immune tolerance, which always causes severe irreversible systematical organ damage and threatens human health heavily. To date, there are still no definitive cures for the treatment of AIDs due to their pathogenesis has not been clearly understood. Besides, the current clinical treatments of AIDs majorly rely on glucocorticoids and immune suppressors, which can lead to serious side effects. In the past years, there are increasing studies demonstrating that an imbalance of gut microbiota is intimately related to the pathogenesis of various AIDs, shedding light on the development of therapeutics by targeting the gut microbiota for the management of AIDs. Among all the approaches targeting the gut microbiota, fecal microbiota transplantation (FMT) has attracted increasing interest, and it has been proposed as a possible strategy to intervene in the homeostasis of gut microbiota for the treatment of various diseases. However, despite the reported good curative effects and clinical studies conducted on FMT, the detailed mechanisms of FMT for the effective treatment of those diseases have not been figured out. To fully understand the mechanisms of the therapeutic effects of FMT on AIDs and improve the therapeutic efficacy of FMT treatment, a systematic review of this topic is necessary. Hence, in this review paper, the potential mechanisms of FMT for the treatment of various AIDs were summarized, including promotion, shaping, activation, or inhibition of the host immune system via the interactions between the microorganisms and the gut immune system, gut-brain, gut-liver, gut-kidney axis, and so on. Then, applications of FMT for the treatment of various AIDs were detailed presented. Finally, the current challenges and potential solutions for the development of FMT formulations and FMT therapeutics were comprehensively discussed.
Collapse
Affiliation(s)
- Xiaomin Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, PR China
| | - Mei Liu
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, PR China
| | - Ming Zhao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, PR China; Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, The Second Xiangya Hospital of Central South University, Changsha, 421142, PR China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, PR China
| | - Changxing Gao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, PR China
| | - Xinyu Fan
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, PR China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, PR China.
| | - Qianjin Lu
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, PR China; Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, The Second Xiangya Hospital of Central South University, Changsha, 421142, PR China.
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, PR China.
| |
Collapse
|
4
|
Yedke NG, Arthur R, Kumar P. Bacillus calmette gaurine vaccine ameliorates the neurotoxicity of quinolinic acid in rats via the modulation of antioxidant, inflammatory and apoptotic markers. J Chem Neuroanat 2023; 131:102287. [PMID: 37172828 DOI: 10.1016/j.jchemneu.2023.102287] [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: 01/25/2023] [Revised: 04/06/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
A mutation in the Huntingtin gene causes 'Huntington's disease, which presents as a motor and behavioral impairment. Due to the limited drug therapy for this disease, scientists are constantly searching for newer and alternative drugs that may either retard or prevent the progress of the disease. This study aims to explore the neuroprotective potential of Bacillus Calmette Gaurine (BCG) vaccine against quinolinic acid-induced (QA) neurotoxicity in rats. QA (200 nmol/2 µl, i.s) was injected bilaterally into the rat striatum, after which a single dose of BCG (2 × 10^7, cfu) was given to the rats. Animals were assessed for behavioral parameters on the 14th and 21st days. On the 22nd day, animals were sacrificed, brains were harvested, and striatum was separated to evaluate biochemical, inflammatory, and apoptotic mediators. Histopathological studies were performed using Hematoxyline and Eosin staining to assess neuronal morphology. BCG treatment reversed motor abnormalities, reduced oxidative stress and neuroinflammatory markers, apoptotic mediators and striatal lesions induced by QA treatment. In conclusion, treat' 'ing rats with BCG vaccine (2 × 10^7, cfu) mitigated the quinolinic acid-induced Huntington's disease-like symptoms. Hence, BCG vaccine (2 ×10^7, cfu) could be used as an adjuvant in managing HD.
Collapse
Affiliation(s)
- Narhari Gangaram Yedke
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda 151001, Punjab, India; Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Richmond Arthur
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| |
Collapse
|
5
|
Plocica J, Guo F, Das JK, Kobayashi KS, Ficht TA, Alaniz RC, Song J, de Figueiredo P. Engineering live attenuated vaccines: Old dogs learning new tricks. J Transl Autoimmun 2023; 6:100198. [PMID: 37090898 PMCID: PMC10113845 DOI: 10.1016/j.jtauto.2023.100198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/24/2023] [Accepted: 03/26/2023] [Indexed: 03/29/2023] Open
Abstract
Autoimmune diseases such as rheumatoid arthritis and type 1 diabetes are increasingly common global problems. Concerns about increases in the prevalence of such diseases and the limited efficacy of conventional treatment regimens necessitates new therapies to address these challenges. Autoimmune disease severity and dysbiosis are interconnected. Although probiotics have been established as a therapy to rebalance the microbiome and suppress autoimmune symptoms, these microbes tend to lack a number of advantageous qualities found in non-commensal bacteria. Through attenuation and genetic manipulation, these non-commensal bacteria have been engineered into recombinant forms that offer malleable platforms capable of addressing the immune imbalances found in RA and T1D. Such bacteria have been engineered to express valuable gene products known to suppress autoimmunity such as anti-inflammatory cytokines, autoantigens, and enzymes synthesizing microbial metabolites. This review will highlight current and emerging trends in the field and discuss how they may be used to prevent and control autoimmune diseases.
Collapse
Affiliation(s)
- Julia Plocica
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Fengguang Guo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Jugal Kishore Das
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Koichi S. Kobayashi
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
- Department of Immunology, Graduate School of Medicine, Hokkaido University Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
- Institute of Vaccine Research and Development, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Thomas A. Ficht
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, 77845, USA
| | - Robert C. Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Paul de Figueiredo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, 77845, USA
| |
Collapse
|
6
|
Chen J, Gao L, Wu X, Fan Y, Liu M, Peng L, Song J, Li B, Liu A, Bao F. BCG-induced trained immunity: history, mechanisms and potential applications. J Transl Med 2023; 21:106. [PMID: 36765373 PMCID: PMC9913021 DOI: 10.1186/s12967-023-03944-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/31/2023] [Indexed: 02/12/2023] Open
Abstract
The Bacillus Calmette-Guérin (BCG) vaccine was discovered a century ago and has since been clinically applicable. BCG can not only be used for the prevention of tuberculosis, but also has a non-specific protective effect on the human body called trained immunity that is mediated by innate immune cells such as monocytes, macrophages, and natural killer cells. Mechanisms of trained immunity include epigenetic reprogramming, metabolic reprogramming, and long-term protection mediated by hematopoietic stem cells. Trained immunity has so far shown beneficial effects on cancer, viral-infections, autoimmune diseases, and a variety of other diseases, especially bladder cancer, respiratory viruses, and type 1 diabetes. The modulation of the immune response by BCG has led to the development of a variety of recombinant vaccines. Although the specific mechanism of BCG prevention on diseases has not been fully clarified, the potential role of BCG deserves further exploration, which is of great significance for prevention and treatment of diseases.
Collapse
Affiliation(s)
- Jingjing Chen
- grid.285847.40000 0000 9588 0960The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500 Yunnan China ,grid.285847.40000 0000 9588 0960Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Li Gao
- grid.285847.40000 0000 9588 0960The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500 Yunnan China ,grid.285847.40000 0000 9588 0960Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Xinya Wu
- grid.285847.40000 0000 9588 0960The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500 Yunnan China ,grid.285847.40000 0000 9588 0960Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Yuxin Fan
- grid.285847.40000 0000 9588 0960The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500 Yunnan China ,grid.285847.40000 0000 9588 0960Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Meixiao Liu
- grid.285847.40000 0000 9588 0960The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500 Yunnan China ,grid.285847.40000 0000 9588 0960Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Li Peng
- grid.285847.40000 0000 9588 0960The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500 Yunnan China ,grid.285847.40000 0000 9588 0960Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Jieqin Song
- grid.285847.40000 0000 9588 0960The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500 Yunnan China ,grid.285847.40000 0000 9588 0960Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Bingxue Li
- grid.285847.40000 0000 9588 0960The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500 Yunnan China ,grid.285847.40000 0000 9588 0960Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Aihua Liu
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China. .,Yunnan Health Cell Biotechnology Company, Kunming, 650041, Yunnan, China. .,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China.
| | - Fukai Bao
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China. .,Yunnan Health Cell Biotechnology Company, Kunming, 650041, Yunnan, China. .,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, Yunnan, China.
| |
Collapse
|
7
|
Senousy MA, Hanafy ME, Shehata N, Rizk SM. Erythropoietin and Bacillus Calmette-Guérin Vaccination Mitigate 3-Nitropropionic Acid-Induced Huntington-like Disease in Rats by Modulating the PI3K/Akt/mTOR/P70S6K Pathway and Enhancing the Autophagy. ACS Chem Neurosci 2022; 13:721-732. [PMID: 35226456 DOI: 10.1021/acschemneuro.1c00523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Oxidative stress and mitochondrial dysfunction are among the mechanisms expected to explain the pathogenesis of Huntington's disease. Erythropoietin (EPO) and the Bacillus Calmette-Guérin (BCG) vaccine have neuroprotective effects against neurodegenerative diseases; however, the full mechanisms of their action are currently unclear. Here, for the first time, we investigated the neuroprotective effect of BCG vaccination in Huntington-like disease induced by 3-nitropropionic acid (3-NP) and its combination with EPO. Male Wistar rats were randomized into five groups: saline-treated control; 3-NP group (20 mg/kg/day, i.p.) for 7 days; EPO-treated group (5000 IU/kg/day, i.p.) for 14 days after 3-NP administration; live BCG vaccine prophylactic group (5000 cfu/g, i.p.) 10 days prior to 3-NP administration; and live BCG vaccine (5000 cfu/g, i.p.) 10 days before 3-NP administration, followed by EPO treatment (5000 IU/kg/day, i.p.) for 14 days. In a histopathological examination, striatum neurodegeneration was evidenced in the 3-NP injected rats. Administration of 3-NP elevated the levels of p-PI3K, p-Akt, p-mTOR, p-P70S6K, BAX, malondialdehyde, nitric oxide, and cytochrome oxidase while reduced the levels of BCL-2, superoxide dismutase, reduced glutathione, and the autophagy marker microtubule-associated protein light chain 3 in the striatum. EPO and BCG ameliorated the biochemical, histopathological, and behavioral derangements induced by 3-NP, with prominent neuroprotection observed in rats administered the BCG prophylactic combined with EPO treatment. These results highlight the role played by EPO and BCG in the management of 3-NP-induced Huntington-like disease by inhibiting the PI3K/Akt/mTOR/P70S6K pathway and enhancing the autophagy.
Collapse
Affiliation(s)
- Mahmoud A. Senousy
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Mona Essam Hanafy
- Central Administration of Biological and Innovative Products and Clinical Studies, Egyptian Drug Authority, Giza 00202, Egypt
| | - Nahla Shehata
- Central Administration of Biological and Innovative Products and Clinical Studies, Egyptian Drug Authority, Giza 00202, Egypt
| | - Sherine M. Rizk
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
8
|
Dow CT, Greenblatt CL, Chan ED, Dow JF. Evaluation of BCG Vaccination and Plasma Amyloid: A Prospective, Pilot Study with Implications for Alzheimer’s Disease. Microorganisms 2022; 10:microorganisms10020424. [PMID: 35208878 PMCID: PMC8880735 DOI: 10.3390/microorganisms10020424] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/04/2022] [Accepted: 02/09/2022] [Indexed: 02/01/2023] Open
Abstract
BCG vaccine has been used for 100 years to prevent tuberculosis. Not all countries, including the United States, adopted the initial World Health Organization recommendation to use BCG. Moreover, many Western countries that had routinely used BCG have discontinued its use. Recent population studies demonstrate lower prevalence of Alzheimer’s disease (AD) in countries with high BCG coverage. Intravesicular instillation of BCG is also used to treat bladder cancer that has not invaded the bladder muscle wall and has been shown to reduce recurrence. Several retrospective studies of bladder cancer patients demonstrated that BCG treatment was associated with a significantly reduced risk of developing AD. Plasma amyloid β assessment has become a fertile area of study for an AD biomarker that is predictive of a positive amyloid PET scan. Mass spectrometry-based plasma amyloid 42/40 ratio has proven to be accurate and robust, and when combined with age and ApoE, is shown to accurately predict current and future brain amyloid status. These parameters, amyloid 42/40 ratio, age and ApoE genotype are incorporated into an Amyloid Probability Score (APS)–a score that identifies low, intermediate or high risk of having a PET scan positive for cerebral amyloid. Community recruitment was used for this open-label pilot study. Forty-nine BCG-naïve, immunocompetent individuals completed our study: prior to BCG prime and boost, as determined by the APS, 34 had low risk (APS 0–35), 5 had intermediate risk (APS 36–57) and 10 had high risk (APS 58–100). The APS range for the participant group was 0 to 94. Follow-up plasma amyloid testing 9 months after vaccination revealed a reduction in the APS in all the risk groups: low risk group (p = 0. 37), intermediate risk group (p = 0.13) and the high-risk group (statistically significant, p = 0.016). Greater benefit was seen in younger participants and those with the highest risk. The small number of participants and the nascent status of plasma amyloid testing will rightfully temper embracement of these results. However, both the favorable direction of change after BCG as well as the utility of the APS—a valuable surrogate AD biomarker—may prompt a definitive large-scale multicenter investigation of BCG and AD risk as determined by plasma amyloid peptide ratios and APS.
Collapse
Affiliation(s)
- Coad Thomas Dow
- Department of Ophthalmology and Visual Sciences, McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
- Mindful Diagnostics and Therapeutics, Eau Claire, WI 54701, USA
- Correspondence:
| | - Charles L. Greenblatt
- Department of Microbiology and Molecular Genetics, Hebrew University, Jerusalem 9103401, Israel;
| | - Edward D. Chan
- Department of Academic Affairs, National Jewish Health, Denver, CO 80218, USA;
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80217, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045, USA
| | - Jordan F. Dow
- Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA;
- Northwestern Wisconsin Region Mayo Clinic Health System, Eau Claire, WI 54703, USA
| |
Collapse
|
9
|
Cossu D, Ruberto S, Yokoyama K, Hattori N, Sechi LA. Efficacy of BCG vaccine in animal models of neurological disorders. Vaccine 2021; 40:432-436. [PMID: 34906393 DOI: 10.1016/j.vaccine.2021.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/02/2021] [Indexed: 10/19/2022]
Abstract
The Bacillus Calmette-Guerin (BCG) vaccine can modulate the immune response via antigen-specific immune response, but also it can confer nonspecific protection and therapeutic benefits in several neurological conditions through different heterologous effects of vaccination. However, the precise mechanism of action of BCG remains unclear. In this review, different mechanisms underlying BCG-mediated immunity will be explained in animal models that reflects characteristic feature of neuroinflammatory and neurodegenerative disorders such as multiple sclerosis, Alzheimer's and Parkinson's diseases. Furthermore, evidence for a beneficial effect of the BCG on neuropsychiatric disorders, will be also discussed.
Collapse
Affiliation(s)
- Davide Cossu
- University of Sassari, Department of Biomedical Sciences, Division of Microbiology and Virology, Sassari 09100, Italy; Juntendo University, Department of Neurology, Tokyo 113-8431, Japan.
| | - Stefano Ruberto
- University of Sassari, Department of Biomedical Sciences, Division of Microbiology and Virology, Sassari 09100, Italy
| | | | - Nobutaka Hattori
- Juntendo University, Department of Neurology, Tokyo 113-8431, Japan
| | - Leonardo A Sechi
- University of Sassari, Department of Biomedical Sciences, Division of Microbiology and Virology, Sassari 09100, Italy; SC Microbiologia AOU Sassari, Sassari, Italy.
| |
Collapse
|
10
|
Matsuzaki G, Teruya N, Kiyohara Kohama H, Arai K, Shibuya Y, Chuma Y, Matsuo K. Mycobacterium bovis BCG-mediated suppression of Th17 response in mouse experimental autoimmune encephalomyelitis. Immunopharmacol Immunotoxicol 2021; 43:203-211. [PMID: 33541144 DOI: 10.1080/08923973.2021.1878215] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is an autoimmune disease mediated by a pro-inflammatory immune response. Experimental autoimmune encephalomyelitis (EAE) induced by immunization of mice with a myelin oligodendrocyte glycoprotein (MOG) peptide emulsified in killed Mycobacterium tuberculosis-containing complete Freund's adjuvant (CFA-EAE) is used as a model of MS. Mycobacterium bovis BCG has been reported to ameliorate clinical symptoms of CFA-EAE, although the precise mechanism has not yet been documented. Since CFA-EAE uses adjuvant with mycobacterial antigens, mycobacterial antigen-specific T cells induced by CFA may cross-react with BCG and modulate EAE. METHODS To exclude the influence of cross-reactivity, a modified murine EAE model (cell wall skeleton (CWS)-EAE) that does not induce mycobacterial antigen-specific T cells was established and used to reevaluate the therapeutic effects of BCG on EAE. RESULTS Inoculation with BCG 6 d after CWS-EAE induction successfully ameliorated EAE symptoms, suggesting that the therapeutic effects of BCG are independent of the mycobacterial antigen-specific T cells induced by the CFA-EAE protocol. BCG inoculation into the CWS-EAE mice resulted in reduced levels of MOG-specific Th17 in the central nervous system (CNS) with reduced demyelinated lesions of the spinal cord. In the draining lymph nodes of the MOG-immunized sites, BCG inoculation resulted in an increase in MOG-specific Th17 and Th1 cells at an early stage of immune response. CONCLUSION The results suggest that BCG inoculation suppresses the Th17 response in the CNS of EAE mice via a mechanism that may involve the suppression of egress of encephalitogenic T cells from lymphoid organs.
Collapse
Affiliation(s)
- Goro Matsuzaki
- Molecular Microbiology Group, Tropical Biosphere Research Center, University of the Ryukyus, Nishihara, Japan.,Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Naoko Teruya
- Molecular Microbiology Group, Tropical Biosphere Research Center, University of the Ryukyus, Nishihara, Japan
| | | | - Keiko Arai
- Research and Development Department, Japan BCG Laboratory, Kiyose, Japan
| | - Yukihiro Shibuya
- Research and Development Department, Japan BCG Laboratory, Kiyose, Japan
| | - Yasushi Chuma
- Research and Development Department, Japan BCG Laboratory, Kiyose, Japan
| | - Kazuhiro Matsuo
- Research and Development Department, Japan BCG Laboratory, Kiyose, Japan
| |
Collapse
|
11
|
Adesanya OA, Uche-Orji CI, Adedeji YA, Joshua JI, Adesola AA, Chukwudike CJ. Expanded Scope of Bacillus Calmette-Guerin (BCG) Vaccine Applicability in Disease Prophylaxis, Diagnostics, and Immunotherapeutics. INFECTIOUS MICROBES & DISEASES 2020; 2:144-150. [PMID: 38630099 PMCID: PMC7769055 DOI: 10.1097/im9.0000000000000040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 11/29/2022]
Abstract
Following the discovery of the Bacillus Calmette-Guerin (BCG) vaccine, its efficacy against Mycobacterium tuberculosis was soon established, with several countries adopting universal BCG vaccination schemes for their populations. Soon, however, studies aimed to further establish the efficacy of the vaccine in different populations discovered that the vaccine has a larger effect in reducing mortality rate than could be explained by its effect on tuberculosis alone, which sparked suggestions that the BCG vaccine could have effects on other unrelated or non-mycobacterial pathogens causing diseases in humans. These effects were termed heterologous, non-specific or off-target effects and have been shown to be due to both innate and adaptive immune system responses. Experiments carried out in a bid to further understand these effects led to many more discoveries about the applicability of the BCG vaccine for the prevention, diagnosis, and treatment of certain disease conditions. As we approach the second century since the discovery of the vaccine, we believe it is timely to review these interesting applications of the BCG vaccine, such as in the prevention of diabetes, atherosclerosis, and leukemia; the diagnosis of Kawasaki disease; and the treatment of multiple sclerosis, non-muscle invading bladder cancer, and stage III melanoma. Furthermore, complications associated with the administration of the BCG vaccine to certain groups of patients, including those with severe combined immunodeficiency and HIV, have been well described in literature, and we conclude by describing the mechanisms behind these complications and discuss their implications on vaccination strategies, especially in low-resource settings.
Collapse
Affiliation(s)
- Oluwafolajimi A. Adesanya
- Institute for Advanced Medical Research and Training (IAMRAT), College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Yeshua A. Adedeji
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - John I. Joshua
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeniyi A. Adesola
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | |
Collapse
|
12
|
Yamazaki-Nakashimada MA, Unzueta A, Berenise Gámez-González L, González-Saldaña N, Sorensen RU. BCG: a vaccine with multiple faces. Hum Vaccin Immunother 2020; 16:1841-1850. [PMID: 31995448 DOI: 10.1080/21645515.2019.1706930] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BCG has been recommended because of its efficacy against disseminated and meningeal tuberculosis. The BCG vaccine has other mechanisms of action besides tuberculosis protection, with immunomodulatory properties that are now being discovered. Reports have shown a significant protective effect against leprosy. Randomized controlled trials suggest that BCG vaccine has beneficial heterologous (nonspecific) effects on mortality in some developing countries. BCG immunotherapy is considered the gold standard adjuvant treatment for non-muscle-invasive bladder cancer. BCG vaccine has also been tested as treatment for diabetes and multiple sclerosis. Erythema of the BCG site is recognized as a clinical clue in Kawasaki disease. BCG administration in the immunodeficient patient is associated with local BCG disease (BCGitis) or disseminated BCG disease (BCGosis) with fatal consequences. BCG administration has been associated with the development of autoimmunity. We present a brief review of the diverse facets of the vaccine, with the discovery of its new modes of action providing new perspectives on this old, multifaceted and controversial vaccine.
Collapse
Affiliation(s)
| | - Alberto Unzueta
- Gastroenterology and Transplant Hepatology, Geisinger Medical Center , Danville, PA, USA
| | | | | | - Ricardo U Sorensen
- Department of Pediatrics, Louisiana State University Health Sciences Center, Louisiana Primary Immunodeficiency Network , New Orleans, LA, USA.,Faculty of Medicine, University of La Frontera , Temuco, Chile
| |
Collapse
|
13
|
Cavallo S. Immune-mediated genesis of multiple sclerosis. J Transl Autoimmun 2020; 3:100039. [PMID: 32743522 PMCID: PMC7388381 DOI: 10.1016/j.jtauto.2020.100039] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/03/2020] [Accepted: 01/05/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is widely acknowledged to be an autoimmune disease affecting the neuronal myelin structure of the CNS. Autoantigens recognized as the target of this autoimmune process are: myelin basal protein, anti-proteolipid protein, antimyelin-associated glycoprotein and antimyelin-based oligodendrocytic basic protein. Ample evidence supports the idea of a dysregulation of immunological tolerance towards self-antigens of neuronal myelin structure triggered by one or more viral or bacterial microbial agents in predisposed HLA gene subjects. Genetic predisposition to MS has been highlighted by numerous studies associating the disease to specific HLA haplotypes. Moreover, a wide range of evidence supports the fact that MS may be consequence of one or more viral or bacterial infections such as measles virus, EBV, HHV6, HZV, Chlamydia pneumoniae, Helicobacter Pylori, and other microbial agents. Microbiota elements also seems to have a role on the determinism of the disease as a pathogenic or protective factor. The autoimmune pathogenetic process could arise when a molecular mimicry between a foreign microbial antigen and an auto-antigen occurs in an HLA gene subject competent for that particular antigen. The antigen-presenting cells in this case would induce the activation of a specific Th clone causing a cross-reaction between a foreign antigen and an autoantigen resulting in an autoimmune response. A multifactorial ethiopathogenetic model based on immunomediation is a reliable hypothesis for multiple sclerosis. Evidence found in the scientific literature makes it possible to reconstruct this etiopathogenetic hypothesis for MS. HLA gene predisposition, correlation with infections, molecular mimicry and other immunological data are reported.
Collapse
Affiliation(s)
- Salvatore Cavallo
- Expert Doctor in Non-Conventional Medicine, Professor and Member of the Board of the MMS, MMS (Medicina di Modulazione Dei Sistemi) Roma, Salvatore Cavallo Via G.B. Pergolesi, 28, 75100, Matera, Italy
| |
Collapse
|
14
|
Gofrit ON, Klein BY, Cohen IR, Ben-Hur T, Greenblatt CL, Bercovier H. Bacillus Calmette-Guérin (BCG) therapy lowers the incidence of Alzheimer's disease in bladder cancer patients. PLoS One 2019; 14:e0224433. [PMID: 31697701 PMCID: PMC6837488 DOI: 10.1371/journal.pone.0224433] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/14/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) affects one in ten people older than 65 years. Thus far, there is no cure or even disease-modifying treatment for this disease. The immune system is a major player in the pathogenesis of AD. Bacillus Calmette-Guérin (BCG), developed as a vaccine against tuberculosis, modulates the immune system and reduces recurrence of non-muscle invasive bladder cancer. Theoretical considerations suggested that treatment with BCG may decrease the risk of AD. We tested this hypothesis on a natural population of bladder cancer patients. METHODS AND FINDINGS After removing all bladder cancer patients presenting with AD or developing AD within one-year following diagnosis of bladder cancer, we collected data on a total of 1371 patients (1134 males and 237 females) who were followed for at least one year after the diagnosis of bladder cancer. The mean age at diagnosis of bladder cancer was 68.1 years (SD 13.0). Adjuvant post-operative intra-vesical treatment with BCG was given to 878 (64%) of these patients. The median period post-operative follow-up was 8 years. During follow-up, 65 patients developed AD at a mean age of 84 years (SD 5.9), including 21 patients (2.4%) who had been treated with BCG and 44 patients (8.9%) who had not received BCG. Patients who had been treated with BCG manifested more than 4-fold less risk for AD than those not treated with BCG. The Cox proportional hazards regression model and the Kaplan-Meier analysis of AD free survival both indicated high significance: patients not treated with BCG had a significantly higher risk of developing AD compared to BCG treated patients (HR 4.778, 95%CI: 2.837-8.046, p = 4.08x10-9 and Log Rank Chi-square 42.438, df = 1, p = 7.30x10-11, respectively). Exposure to BCG did not modify the prevalence of Parkinson's disease, 1.9% in BCG treated patients and 1.6% in untreated (Fisher's Exact Test, p = 1). CONCLUSIONS Bladder cancer patients treated with BCG were significantly less likely to develop AD at any age than patients who were not so treated. This finding of a retrospective study suggests that BCG treatment might also reduce the incidence of AD in the general population. Confirmation of such effects of BCG in other retrospective studies would support prospective studies of BCG in AD.
Collapse
Affiliation(s)
- Ofer N. Gofrit
- Department of Urology, Hadassah- Hebrew University Medical Center, Jerusalem, Israel
- * E-mail: (HB); (ONG)
| | - Benjamin Y. Klein
- Department of Microbiology and Molecular Genetics, Hebrew University Jerusalem, Israel
| | - Irun R. Cohen
- Department of Immunology, Weizmann Institute, Rehovot, Israel
| | - Tamir Ben-Hur
- Department of Neurology Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Charles L. Greenblatt
- Department of Microbiology and Molecular Genetics, Hebrew University Jerusalem, Israel
| | - Hervé Bercovier
- Department of Microbiology and Molecular Genetics, Hebrew University Jerusalem, Israel
- * E-mail: (HB); (ONG)
| |
Collapse
|
15
|
Usher NT, Chang S, Howard RS, Martinez A, Harrison LH, Santosham M, Aronson NE. Association of BCG Vaccination in Childhood With Subsequent Cancer Diagnoses: A 60-Year Follow-up of a Clinical Trial. JAMA Netw Open 2019; 2:e1912014. [PMID: 31553471 PMCID: PMC6763973 DOI: 10.1001/jamanetworkopen.2019.12014] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IMPORTANCE The BCG vaccine is currently the only approved tuberculosis vaccine and is widely administered worldwide, usually during infancy. Previous studies found increased rates of lymphoma and leukemia in BCG-vaccinated populations. OBJECTIVE To determine whether BCG vaccination was associated with cancer rates in a secondary analysis of a BCG vaccine trial. DESIGN, SETTING, AND PARTICIPANTS Retrospective review (60-year follow-up) of a clinical trial in which participants were assigned to the vaccine group by systematic stratification by school district, age, and sex, then randomized by alternation. The original study was conducted at 9 sites in 5 US states between December 1935 and December 1998. Participants were 2963 American Indian and Alaska Native schoolchildren younger than 20 years with no evidence of previous tuberculosis infection. Statistical analysis was conducted between August 2018 and July 2019. INTERVENTIONS Single intradermal injection of either BCG vaccine or saline placebo. MAIN OUTCOMES AND MEASURES The primary outcome was diagnosis of cancer after BCG vaccination. Data on participant interval health and risk factors, including smoking, tuberculosis infection, isoniazid use, and other basic demographic information, were also collected. RESULTS A total of 2963 participants, including 1540 in the BCG vaccine group and 1423 in the placebo group, remained after exclusions. Vaccination occurred at a median (interquartile range) age of 8 (5-11) years; 805 participants (52%) in the BCG group and 710 (50%) in the placebo group were female. At the time of follow-up, 97 participants (7%) in the placebo group and 106 participants (7%) in the BCG vaccine group could not be located; total mortality was 633 participants (44%) in the placebo group and 632 participants (41%) in the BCG group. The overall rate of cancer diagnosis was not significantly different in BCG vaccine vs placebo recipients (hazard ratio, 0.82; 95% CI, 0.66-1.02), including for lymphoma and leukemia. The rate of lung cancer was significantly lower in BCG vs placebo recipients (18.2 vs 45.4 cases per 100 000 person-years; hazard ratio, 0.38; 95% CI, 0.20-0.74; P = .005), controlling for sex, region, alcohol overuse, smoking, and tuberculosis. CONCLUSIONS AND RELEVANCE Childhood BCG vaccination was associated with a lower risk of lung cancer development in American Indian and Alaska Native populations. This finding has potentially important health implications given the high mortality rate associated with lung cancer and the availability of low-cost BCG vaccines.
Collapse
Affiliation(s)
- Nicholas T. Usher
- Infectious Diseases Division, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- College of Agriculture and Life Sciences, Cornell University, Ithaca, New York
| | - Suyoung Chang
- Division of Vaccines and Related Product Applications, US Food and Drug Administration, Silver Spring, Maryland
| | - Robin S. Howard
- Department of Research Programs, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Adriana Martinez
- Department of Research Programs, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Lee H. Harrison
- Infectious Diseases Epidemiology Research Unit, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mathuram Santosham
- Health Systems Program, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
- Center for American Indian Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Naomi E. Aronson
- Infectious Diseases Division, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
16
|
Tanner R, Villarreal-Ramos B, Vordermeier HM, McShane H. The Humoral Immune Response to BCG Vaccination. Front Immunol 2019; 10:1317. [PMID: 31244856 PMCID: PMC6579862 DOI: 10.3389/fimmu.2019.01317] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/23/2019] [Indexed: 01/19/2023] Open
Abstract
Bacillus Calmette Guérin (BCG) is the only currently available vaccine against tuberculosis (TB), but it confers incomplete and variable protection against pulmonary TB in humans and bovine TB (bTB) in cattle. Insights into the immune response induced by BCG offer an underexploited opportunity to gain knowledge that may inform the design of a more efficacious vaccine, which is urgently needed to control these major global epidemics. Humoral immunity in TB and bTB has been neglected, but recent studies supporting a role for antibodies in protection against TB has driven a growing interest in determining their relevance to vaccine development. In this manuscript we review what is known about the humoral immune response to BCG vaccination and re-vaccination across species, including evidence for the induction of specific B cells and antibodies; and how these may relate to protection from TB or bTB. We discuss potential explanations for often conflicting findings and consider how factors such as BCG strain, manufacturing methodology and route of administration influence the humoral response. As novel vaccination strategies include BCG prime-boost regimens, the literature regarding off-target immunomodulatory effects of BCG vaccination on non-specific humoral immunity is also reviewed. Overall, reported outcomes to date are inconsistent, but indicate that humoral responses are heterogeneous and may play different roles in different species, populations, or individual hosts. Further study is warranted to determine whether a new TB vaccine could benefit from the targeting of humoral as well as cell-mediated immunity.
Collapse
Affiliation(s)
- Rachel Tanner
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Bernardo Villarreal-Ramos
- Department of Bacteriology, Animal and Plant Health Agency, Addlestone, United Kingdom
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - H. Martin Vordermeier
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Department of Bacteriology, Animal and Plant Health Agency, Addlestone, United Kingdom
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - Helen McShane
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Gofrit ON, Bercovier H, Klein BY, Cohen IR, Ben-Hur T, Greenblatt CL. Can immunization with Bacillus Calmette-Guérin (BCG) protect against Alzheimer’s disease? Med Hypotheses 2019; 123:95-97. [DOI: 10.1016/j.mehy.2019.01.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/24/2018] [Accepted: 01/10/2019] [Indexed: 12/16/2022]
|
18
|
Cossu D, Yokoyama K, Hattori N. Bacteria-Host Interactions in Multiple Sclerosis. Front Microbiol 2018; 9:2966. [PMID: 30564215 PMCID: PMC6288311 DOI: 10.3389/fmicb.2018.02966] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/18/2018] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is caused by a complex interaction of genetic and environmental factors. Numerous causative factors have been identified that play a role in MS, including exposure to bacteria. Mycobacteria, Chlamydia pneumoniae, Helicobacter pylori, and other bacteria have been proposed as risk factors for MS with different mechanisms of action. Conversely, some pathogens may have a protective effect on its etiology. In terms of acquired immunity, molecular mimicry has been hypothesized as the mechanism by which bacterial structures such as DNA, the cell wall, and intracytoplasmic components can activate autoreactive T cells or produce autoantibodies in certain host genetic backgrounds of susceptible individuals. In innate immunity, Toll-like receptors play an essential role in combating invading bacteria, and their activation leads to the release of cytokines or chemokines that mediate effective adaptive immune responses. These receptors may also be involved in central nervous system autoimmunity, and their contribution depends on the infection site and on the pathogen. We have reviewed the current knowledge of the influence of bacteria on MS development, emphasizing the potential mechanisms of action by which bacteria affect MS initiation and/or progression.
Collapse
Affiliation(s)
- Davide Cossu
- Department of Neurology, Juntendo University, Tokyo, Japan.,Advanced Research Institute for Health Science, Juntendo University, Tokyo, Japan
| | - Kazumasa Yokoyama
- Department of Neurology, Juntendo University, Tokyo, Japan.,Advanced Research Institute for Health Science, Juntendo University, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University, Tokyo, Japan.,Advanced Research Institute for Health Science, Juntendo University, Tokyo, Japan
| |
Collapse
|
19
|
The Mycobacterial Adjuvant Analogue TDB Attenuates Neuroinflammation via Mincle-Independent PLC-γ1/PKC/ERK Signaling and Microglial Polarization. Mol Neurobiol 2018; 56:1167-1187. [PMID: 29876879 DOI: 10.1007/s12035-018-1135-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/18/2018] [Indexed: 02/06/2023]
Abstract
Microglial activation has long been recognized as a hallmark of neuroinflammation. Recently, the bacillus Calmette-Guerin (BCG) vaccine has been reported to exert neuroprotective effects against several neurodegenerative disorders. Trehalose-6,6'-dibehenate (TDB) is a synthetic analogue of trehalose-6,6'-dimycolate (TDM, also known as the mycobacterial cord factor) and is a new adjuvant of tuberculosis subunit vaccine currently in clinical trials. Both TDM and TDB can activate macrophages and dendritic cells through binding to C-type lectin receptor Mincle; however, its action mechanism in microglia and their relationship with neuroinflammation are still unknown. In this article, we found that TDB inhibited LPS-induced M1 microglial polarization in primary microglia and BV-2 cells. However, TDB itself had no effects on IKK, p38, and JNK activities or cytokine expression. In contrast, TDB activated ERK1/2 through PLC-γ1/PKC signaling and in turn decreased LPS-induced NF-κB nuclear translocation. Furthermore, TDB-induced AMPK activation via PLC-γ1/calcium/CaMKKβ-dependent pathway and thereby enhanced M2 gene expressions. Interestingly, knocking out Mincle did not alter the anti-inflammatory and M2 polarization effects of TDB in microglia. Conditional media from LPS-stimulated microglial cells can induce in vitro neurotoxicity, and this action was attenuated by TDB. Using a mouse neuroinflammation model, we found that TDB suppressed LPS-induced M1 microglial activation and sickness behavior, but promoted M2 microglial polarization in both WT and Mincle-/- mice. Taken together, our results suggest that TDB can act independently of Mincle to inhibit LPS-induced inflammatory response through PLC-γ1/PKC/ERK signaling and promote microglial polarization towards M2 phenotype via PLC-γ1/calcium/CaMKKβ/AMPK pathway. Thus, TDB may be a promising therapeutic agent for the treatment of neuroinflammatory diseases.
Collapse
|
20
|
Zhong X, Zhou Y, Lu T, Wang Z, Fang L, Peng L, Kermode AG, Qiu W. Infections in neuromyelitis optica spectrum disorder. J Clin Neurosci 2017; 47:14-19. [PMID: 29066232 DOI: 10.1016/j.jocn.2017.10.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/01/2017] [Indexed: 12/19/2022]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory astrocytopathy that has both genetic and environmental causes. A growing body of evidence suggests that the presence of several infectious agents correlates with the development of NMOSD. In this review, we summarize studies that either support or present evidence against the hypothesized association between infection and NMOSD. We will also present an overview of potential mechanisms underlying the pathogenesis of NMOSD. Finally, we provide some beneficial properties that infectious elements may have based on "hygiene hypothesis". It is of great clinical significance to further investigate the complex mechanisms by which infections may affect autoimmune diseases to develop better strategies to prevent and treat them, although so far no causal link between infectious agents and NMOSD has been established.
Collapse
Affiliation(s)
- Xiaonan Zhong
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yifan Zhou
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tingting Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhanhang Wang
- Department of Neurology, Guangdong 999 Brain Hospital, China
| | - Ling Fang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lisheng Peng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Allan G Kermode
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Department of Neurology, Sir Charles Gairdner Hospital, Queen Elizabeth II Medical Centre, Perth, Australia; Institute of Immunology and Infectious Diseases, Murdoch University, Perth, Australia
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
21
|
The hygiene hypothesis in autoimmunity: the role of pathogens and commensals. Nat Rev Immunol 2017; 18:105-120. [PMID: 29034905 DOI: 10.1038/nri.2017.111] [Citation(s) in RCA: 322] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The incidence of autoimmune diseases has been steadily rising. Concomitantly, the incidence of most infectious diseases has declined. This observation gave rise to the hygiene hypothesis, which postulates that a reduction in the frequency of infections contributes directly to the increase in the frequency of autoimmune and allergic diseases. This hypothesis is supported by robust epidemiological data, but the underlying mechanisms are unclear. Pathogens are known to be important, as autoimmune disease is prevented in various experimental models by infection with different bacteria, viruses and parasites. Gut commensal bacteria also play an important role: dysbiosis of the gut flora is observed in patients with autoimmune diseases, although the causal relationship with the occurrence of autoimmune diseases has not been established. Both pathogens and commensals act by stimulating immunoregulatory pathways. Here, I discuss the importance of innate immune receptors, in particular Toll-like receptors, in mediating the protective effect of pathogens and commensals on autoimmunity.
Collapse
|
22
|
Kowalewicz-Kulbat M, Locht C. BCG and protection against inflammatory and auto-immune diseases. Expert Rev Vaccines 2017; 16:1-10. [PMID: 28532186 DOI: 10.1080/14760584.2017.1333906] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Bacillus Calmette-Guérin (BCG) is the only available vaccine against tuberculosis. Although its protective efficacy against pulmonary tuberculosis is still under debate, it provides protection against other mycobacterial diseases. BCG is also an effective therapy against superficial bladder cancer and potentially decreases overall childhood mortality. Areas covered: The purpose of this paper is to provide a state-of-the-art summary of the beneficial effects of BCG in inflammatory and auto-immune diseases. As a strong inducer of Th1 type immunity, BCG has been reported to protect against atopic conditions, such as allergic asthma, a Th2-driven disorder. Its protective effect has been well documented in mice, but still awaits definitive evidence in humans. Similarly, murine studies have shown a protective effect of BCG against auto-immune diseases, such as multiple sclerosis and insulin-dependent diabetes, but studies in humans have come to conflicting conclusions. Expert commentary: Studies in mice have shown a beneficial effect of the BCG vaccine against allergic asthma, multiple sclerosis and diabetes. However, the understanding of its mechanism is still fragmentary and requires further in depth research. Some observational or intervention studies in humans have also suggested a beneficial effect, but definitive evidence for this requires confirmation in carefully conducted prospective studies.
Collapse
Affiliation(s)
- Magdalena Kowalewicz-Kulbat
- a Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology , University of Lodz , Lodz , Poland
| | - Camille Locht
- a Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology , University of Lodz , Lodz , Poland.,b Center for Infection and Immunity of Lille , Institut Pasteur de Lille , Lille , France.,c Center for Infection and Immunity of Lille , Inserm U1019 , Lille , France.,d Center for Infection and Immunity of Lille , CNRS UMR 8204 , Lille , France.,e Center for Infection and Immunity of Lille , Université Lille Nord de France , Lille , France
| |
Collapse
|
23
|
Cossu D, Yokoyama K, Hattori N. Conflicting Role of Mycobacterium Species in Multiple Sclerosis. Front Neurol 2017; 8:216. [PMID: 28579973 PMCID: PMC5437105 DOI: 10.3389/fneur.2017.00216] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/02/2017] [Indexed: 01/05/2023] Open
Abstract
Mycobacterium is a genus of aerobic and acid-fast bacteria, which include several pathogenic organisms that cause serious diseases in mammals. Previous studies have associated the immune response against mycobacteria with multiple sclerosis (MS), a chronic demyelinating disease of the central nervous system with unknown etiology. The role of mycobacteria in the pathological process has been controversial and often conflicting. We provide a detailed review of the mycobacteria that have been linked to MS over the last three decades, with a focus on Mycobacterium bovis bacille Calmette-Guérin vaccine for human and oral exposure to Mycobacterium avium subsp. paratuberculosis. We will also discuss the exposure and genetic susceptibility to mycobacterial infection, the protective role of vaccination, as well as the possible mechanisms involved in initiating or worsening MS symptoms, with particular emphasis on the molecular mimicry between mycobacterial and human proteins. Finally, we will introduce topics such as heat shock proteins and recognition by innate immunity, and toll-like receptor signaling-mediated responses to Mycobacterium exposure.
Collapse
|
24
|
Hsu NJ, Francisco NM, Keeton R, Allie N, Quesniaux VFJ, Ryffel B, Jacobs M. Myeloid and T Cell-Derived TNF Protects against Central Nervous System Tuberculosis. Front Immunol 2017; 8:180. [PMID: 28280495 PMCID: PMC5322283 DOI: 10.3389/fimmu.2017.00180] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 02/07/2017] [Indexed: 01/25/2023] Open
Abstract
Tuberculosis of the central nervous system (CNS-TB) is a devastating complication of tuberculosis, and tumor necrosis factor (TNF) is crucial for innate immunity and controlling the infection. TNF is produced by many cell types upon activation, in particularly the myeloid and T cells during neuroinflammation. Here we used mice with TNF ablation targeted to myeloid and T cell (MT-TNF-/-) to assess the contribution of myeloid and T cell-derived TNF in immune responses during CNS-TB. These mice exhibited impaired innate immunity and high susceptibility to cerebral Mycobacterium tuberculosis infection, a similar phenotype to complete TNF-deficient mice. Further, MT-TNF-/- mice were not able to control T cell responses and cytokine/chemokine production. Thus, our data suggested that collective TNF production by both myeloid and T cells are required to provide overall protective immunity against CNS-TB infection.
Collapse
Affiliation(s)
- Nai-Jen Hsu
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town , Cape Town , South Africa
| | - Ngiambudulu M Francisco
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town , Cape Town , South Africa
| | - Roanne Keeton
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town , Cape Town , South Africa
| | - Nasiema Allie
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town , Cape Town , South Africa
| | - Valérie F J Quesniaux
- CNRS UMR7355, Experimental and Molecular Immunology and Neurogenetics , Orleans , France
| | - Bernhard Ryffel
- CNRS UMR7355, Experimental and Molecular Immunology and Neurogenetics , Orleans , France
| | - Muazzam Jacobs
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; South African Medical Research Council, Cape Town, South Africa; National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
25
|
Yang J, Qi F, Yang Y, Yuan Q, Zou J, Guo K, Yao Z. Neonatal hepatitis B vaccination impaired the behavior and neurogenesis of mice transiently in early adulthood. Psychoneuroendocrinology 2016; 73:166-176. [PMID: 27501128 DOI: 10.1016/j.psyneuen.2016.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 07/09/2016] [Accepted: 08/01/2016] [Indexed: 01/01/2023]
Abstract
The immune system plays a vital role in brain development. The hepatitis B vaccine (HBV) is administered to more than 70% of neonates worldwide. Whether this neonatal vaccination affects brain development is unknown. Newborn C57BL/6 mice were injected intraperitoneally with HBV or phosphate-buffered saline. HBV induced impaired behavioral performances and hippocampal long-term potentiation at 8 weeks (w) of age without influence at 4 or 12w. At 6w, there was decreased neurogenesis, M1 microglial activation and a neurotoxic profile of neuroimmune molecule expression [increased tumor necrosis factor-α and reduced interferon (IFN)-γ, brain-derived neurotrophic factor and insulin-like growth factor-1] in the hippocampus of the HBV-vaccinated mice. In the serum, HBV induced significantly higher levels of interleukin (IL)-4, indicating a T helper (Th)-2 bias. Moreover, the serum IFN-γ/IL-4 ratio was positively correlated with the levels of neurotrophins and neurogenesis in the hippocampus at the individual level. These findings suggest that neonatal HBV vaccination of mice results in neurobehavioral impairments in early adulthood by inducing a proinflammatory and low neurotrophic milieu in the hippocampus, which follows the HBV-induced systemic Th2 bias.
Collapse
Affiliation(s)
- Junhua Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Yang Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Qunfang Yuan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Juntao Zou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Kaihua Guo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China.
| |
Collapse
|
26
|
Flanagan KL, Plebanski M. Sex-differential heterologous (non-specific) effects of vaccines: an emerging public health issue that needs to be understood and exploited. Expert Rev Vaccines 2016; 16:5-13. [DOI: 10.1080/14760584.2016.1203260] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Katie L. Flanagan
- Vaccine and Infectious Diseases Laboratory, Department of Immunology and Pathology, Monash University, Prahran, Australia
| | - Magdalena Plebanski
- Vaccine and Infectious Diseases Laboratory, Department of Immunology and Pathology, Monash University, Prahran, Australia
- Monash Institute of Medical Engineering, Monash University, Prahran, Australia
| |
Collapse
|
27
|
Neonatal Bacillus Calmette-Guérin vaccination alleviates lipopolysaccharide-induced neurobehavioral impairments and neuroinflammation in adult mice. Mol Med Rep 2016; 14:1574-86. [PMID: 27357155 PMCID: PMC4940080 DOI: 10.3892/mmr.2016.5425] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 04/29/2016] [Indexed: 12/23/2022] Open
Abstract
The Bacillus Calmette-Guérin (BCG) vaccine is routinely administered to human neonates worldwide. BCG has recently been identified as a neuroprotective immune mediator in several neuropathological conditions, exerting neuroprotection in a mouse model of Parkinson's disease and slowing the progression of clinically isolated syndrome in patients with multiple sclerosis. The immune system is significantly involved in brain development, and several types of neonatal immune activations exert influences on the brain and behavior following a secondary immune challenge in adulthood. However, whether the neonatal BCG vaccination affects the brain in adulthood remains to be elucidated. In the present study, newborn C57BL/6 mice were injected subcutaneously with BCG (105 colony forming units) or phosphate-buffered saline (PBS). A total of 12 weeks later, the mice were injected intraperitoneally with 330 µg/kg lipopolysaccharide (LPS) or PBS. The present study reported that the neonatal BCG vaccination alleviated sickness, anxiety and depression-like behavior, lessened the impairments in hippocampal cell proliferation and downregulated the proinflammatory responses in the serum and brain that were induced by the adult LPS challenge. However, BCG vaccination alone had no evident influence on the brain and behavior in adulthood. In conclusion, the neonatal BCG vaccination alleviated the neurobehavioral impairments and neuroinflammation induced by LPS exposure in adult mice, suggesting a potential neuroprotective role of the neonatal BCG vaccination in adulthood.
Collapse
|
28
|
Lee JE, Walsh MC, Hoehn KL, James DE, Wherry EJ, Choi Y. Acetyl CoA Carboxylase 2 Is Dispensable for CD8+ T Cell Responses. PLoS One 2015; 10:e0137776. [PMID: 26367121 PMCID: PMC4569334 DOI: 10.1371/journal.pone.0137776] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 08/21/2015] [Indexed: 01/17/2023] Open
Abstract
Differentiation of T cells is closely associated with dynamic changes in nutrient and energy metabolism. However, the extent to which specific metabolic pathways and molecular components are determinative of CD8+ T cell fate remains unclear. It has been previously established in various tissues that acetyl CoA carboxylase 2 (ACC2) regulates fatty acid oxidation (FAO) by inhibiting carnitine palmitoyltransferase 1 (CPT1), a rate-limiting enzyme of FAO in mitochondria. Here, we explore the cell-intrinsic role of ACC2 in T cell immunity in response to infections. We report here that ACC2 deficiency results in a marginal increase of cellular FAO in CD8+ T cells, but does not appear to influence antigen-specific effector and memory CD8+ T cell responses during infection with listeria or lymphocytic choriomeningitis virus. These results suggest that ACC2 is dispensable for CD8+ T cell responses.
Collapse
Affiliation(s)
- Jang Eun Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - Matthew C. Walsh
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - Kyle L. Hoehn
- Department of Pharmacology, University of Virginia Health System, Charlottesville, Virginia, 22908, United States of America
| | - David E. James
- Charles Perkins Centre Research and Education Hub, University of Sydney, Sydney, New South Wales, Australia
| | - E. John Wherry
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
- * E-mail:
| |
Collapse
|
29
|
Choi HG, Kim WS, Back YW, Kim H, Kwon KW, Kim JS, Shin SJ, Kim HJ. Mycobacterium tuberculosis RpfE promotes simultaneous Th1- and Th17-type T-cell immunity via TLR4-dependent maturation of dendritic cells. Eur J Immunol 2015; 45:1957-71. [PMID: 25907170 DOI: 10.1002/eji.201445329] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/31/2015] [Accepted: 04/21/2015] [Indexed: 11/12/2022]
Abstract
Reciprocal induction of the Th1 and Th17 immune responses is essential for optimal protection against Mycobacterium tuberculosis (Mtb); however, only a few Mtb antigens are known to fulfill this task. A functional role for resuscitation-promoting factor (Rpf) E, a latency-associated member of the Rpf family, in promoting naïve CD4(+) T-cell differentiation toward both Th1 and Th17 cell fates through interaction with dendritic cells (DCs) was identified in this study. RpfE induces DC maturation by increasing expression of surface molecules and the production of IL-6, IL-1β, IL-23p19, IL-12p70, and TNF-α but not IL-10. This induction is mediated through TLR4 binding and subsequent activation of ERK, p38 MAPKs, and NF-κB signaling. RpfE-treated DCs effectively caused naïve CD4(+) T cells to secrete IFN-γ, IL-2, and IL-17A, which resulted in reciprocal expansions of the Th1 and Th17 cell response along with activation of T-bet and RORγt but not GATA-3. Furthermore, lung and spleen cells from Mtb-infected WT mice but not from TLR4(-/-) mice exhibited Th1 and Th17 polarization upon RpfE stimulation. Taken together, our data suggest that RpfE has the potential to be an effective Mtb vaccine because of its ability to activate DCs that simultaneously induce both Th1- and Th17-polarized T-cell expansion.
Collapse
Affiliation(s)
- Han-Gyu Choi
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea.,Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Woo Sik Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Yong Woo Back
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong-Seok Kim
- Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Hwa-Jung Kim
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea.,Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
30
|
Boer MC, Joosten SA, Ottenhoff THM. Regulatory T-Cells at the Interface between Human Host and Pathogens in Infectious Diseases and Vaccination. Front Immunol 2015; 6:217. [PMID: 26029205 PMCID: PMC4426762 DOI: 10.3389/fimmu.2015.00217] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/20/2015] [Indexed: 12/20/2022] Open
Abstract
Regulatory T-cells (Tregs) act at the interface of host and pathogen interactions in human infectious diseases. Tregs are induced by a wide range of pathogens, but distinct effects of Tregs have been demonstrated for different pathogens and in different stages of infection. Moreover, Tregs that are induced by a specific pathogen may non-specifically suppress immunity against other microbes and parasites. Thus, Treg effects need to be assessed not only in homologous but also in heterologous infections and vaccinations. Though Tregs protect the human host against excessive inflammation, they probably also increase the risk of pathogen persistence and chronic disease, and the possibility of disease reactivation later in life. Mycobacterium leprae and Mycobacterium tuberculosis, causing leprosy and tuberculosis, respectively, are among the most ancient microbes known to mankind, and are master manipulators of the immune system toward tolerance and pathogen persistence. The majority of mycobacterial infections occur in settings co-endemic for viral, parasitic, and (other) bacterial coinfections. In this paper, we discuss recent insights in the activation and activity of Tregs in human infectious diseases, with emphasis on early, late, and non-specific effects in disease, coinfections, and vaccination. We highlight mycobacterial infections as important models of modulation of host responses and vaccine-induced immunity by Tregs.
Collapse
Affiliation(s)
- Mardi C Boer
- Department of Infectious Diseases, Leiden University Medical Center , Leiden , Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center , Leiden , Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|
31
|
Cossu D, Mameli G, Masala S, Cocco E, Frau J, Marrosu MG, Sechi LA. Evaluation of the humoral response against mycobacterial peptides, homologous to MOG₃₅₋₅₅, in multiple sclerosis patients. J Neurol Sci 2014; 347:78-81. [PMID: 25271190 DOI: 10.1016/j.jns.2014.09.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/02/2014] [Accepted: 09/15/2014] [Indexed: 02/07/2023]
Abstract
Bacillus Calmette-Guérin (BCG) and Mycobacterium avium subspecies paratuberculosis (MAP) have been associated with multiple sclerosis (MS). Clinical data indicates that BCG vaccination exerts anti-inflammatory effects in MS; conversely, MAP is thought to be one of the possible infectious factors responsible of MS through a molecular mimicry mechanism. A peptide-based indirect ELISA was used to detect antibodies against the encephalitogenic myelin oligodendrocyte glycoprotein (MOG)35-55 epitope, and two mycobacterial peptides sharing sequence homology with the latter: MAP_2619c352-361/BCG_1224355-364 and BCG_3329c64-74. Among 40 MS patients and 39 healthy volunteers included in the study, only MOG35-55 was capable of inducing a significantly higher humoral response in MS subjects compared to controls. Indeed, 11 out of 40 MS subjects (27.5%) and only 2 out of 39 controls (5%) were antibody-positive for MOG35-55 (p=0.01, AUC=0.65). These findings strengthen the importance of MOG35-55 in MS pathogenesis. The MAP and BCG MOG-homologues epitopes investigated were not recognized in MS patients. Overall, the results allow us concluding that sharing homology of linear epitopes is necessary but not sufficient to induce antibody-mediated cross-reactivity.
Collapse
Affiliation(s)
- Davide Cossu
- Department of Biomedical Sciences, Section of Experimental and Clinical Microbiology, University of Sassari, Italy
| | - Giuseppe Mameli
- Department of Biomedical Sciences, Section of Experimental and Clinical Microbiology, University of Sassari, Italy
| | - Speranza Masala
- Department of Biomedical Sciences, Section of Experimental and Clinical Microbiology, University of Sassari, Italy
| | - Eleonora Cocco
- Multiple Sclerosis Centre, Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, Italy
| | - Jessica Frau
- Multiple Sclerosis Centre, Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, Italy
| | - Maria Giovanna Marrosu
- Multiple Sclerosis Centre, Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, Italy
| | - Leonardo Antonio Sechi
- Department of Biomedical Sciences, Section of Experimental and Clinical Microbiology, University of Sassari, Italy.
| |
Collapse
|
32
|
Fragoso YD, Adoni T, Anacleto A, Brooks JBB, Carvalho MDJ, Claudino R, Damasceno A, Ferreira MLB, Gama PDD, Goncalves MVM, Grzesiuk AK, Matta APDC, Parolin MFK. How do we manage and treat a patient with multiple sclerosis at risk of tuberculosis? Expert Rev Neurother 2014; 14:1251-60. [PMID: 25242167 DOI: 10.1586/14737175.2014.962517] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Tuberculosis continues to be a serious health problem worldwide. The disease continues to be underdiagnosed and not properly treated. In conditions that affect the immune system, such as multiple sclerosis (MS), latent tuberculosis may thrive and reactivate during the use of immunomodulatory and immunosuppressive drugs. Among the best treatment options for patients with latent or active tuberculosis who have MS are IFN-β, glatiramer acetate and mitoxantrone. Drugs leading to a reduced number and/or function of lymphocytes should be avoided or used with caution. Tuberculosis must always be investigated in patients with MS and treated with rigor.
Collapse
Affiliation(s)
- Yara Dadalti Fragoso
- Department of Neurology and MS Reference Center, Universidade Metropolitana de Santos, SP, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Although 'self-nonself' and 'danger' theories have improved our understanding of the immune system, successful immunotherapy of cancer and many autoimmune diseases still remain far from reach. This indicates that our knowledge of how the immune system decides to respond effectively or ineffectively is limited. Emerging evidence suggest that decision-making during the immune response is not solely determined by 'nonself' entity of the antigen or damage-associated 'danger' signals. This article provides an overview of the 'self-nonself' and 'danger' models, and suggests that 'adaptation' signals are needed to guarantee immunological tolerance that has been observed during the immune response toward 'self', 'nonself' or even 'danger'. This should be facilitated by dynamic expression of adapting receptors (ARs) and adapting ligands on cells of the immune system and other somatic cells. Any alterations in the expression of ARs on certain tissues would result in tissue-specific autoimmune diseases or spontaneous regression of cancer. Identification of such ARs and their nominal adapting ligands could lead to the discovery of currently unknown receptors and their implications in the treatment of cancer, solid organ transplantation and autoimmune diseases.
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Box 980035, 401 College Street, Richmond, VA 23298, USA.
| |
Collapse
|
34
|
Andrieu JM, Chen S, Lai C, Guo W, Lu W. Mucosal SIV Vaccines Comprising Inactivated Virus Particles and Bacterial Adjuvants Induce CD8(+) T-Regulatory Cells that Suppress SIV-Positive CD4(+) T-Cell Activation and Prevent SIV Infection in the Macaque Model. Front Immunol 2014; 5:297. [PMID: 25071760 PMCID: PMC4074992 DOI: 10.3389/fimmu.2014.00297] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 06/11/2014] [Indexed: 01/12/2023] Open
Abstract
A new paradigm of mucosal vaccination against human immunodeficiency virus (HIV) infection has been investigated in the macaque model. A vaccine consisting of inactivated simian immunodeficiency virus (SIV)mac239 particles together with a living bacterial adjuvant (either the Calmette and Guerin bacillus, Lactobacillus plantarum or Lactobacillus rhamnosus) was administered to macaques via the vaginal or oral/intragastric route. In contrast to all established human and veterinary vaccines, these three vaccine regimens did not elicit SIV-specific antibodies nor cytotoxic T-lymphocytes but induced a previously unrecognized population of non-cytolytic MHCIb/E-restricted CD8(+) T-regulatory cells that suppressed the activation of SIV-positive CD4(+) T-lymphocytes. SIV reverse transcription was thereby blocked in inactivated CD4(+) T-cells; the initial burst of virus replication was prevented and the vaccinated macaques were protected from a challenge infection. For 3-14 months after intragastric immunization, 24 macaques were challenged intrarectally with a high dose of SIVmac239 or with the heterologous strain SIV B670 (both strains grown on macaques PBMC). Twenty-three of these animals were found to be protected for up to 48 months while all 24 control macaques became infected. This protective effect against SIV challenge together with the concomitant identification of a robust ex vivo correlate of protection suggests a new approach for developing an HIV vaccine in humans. The induction of this new class of CD8(+) T-regulatory cells could also possibly be used therapeutically for suppressing HIV replication in infected patients and this novel tolerogenic vaccine paradigm may have potential applications for treating a wide range of immune disorders and is likely to may have profound implications across immunology generally.
Collapse
Affiliation(s)
- Jean-Marie Andrieu
- Institut de Recherches sur les Vaccins et l'Immunothérapie des Cancers et du SIDA, Centre Universitaire des Saints Peres, Université de Paris-Descartes , Paris , France
| | - Song Chen
- Tropical Medicine Institute, Guangzhou University of Chinese Medicine , Guangzhou , China
| | - Chunhui Lai
- Tropical Medicine Institute, Guangzhou University of Chinese Medicine , Guangzhou , China
| | - Weizhong Guo
- Tropical Medicine Institute, Guangzhou University of Chinese Medicine , Guangzhou , China
| | - Wei Lu
- Institut de Recherches sur les Vaccins et l'Immunothérapie des Cancers et du SIDA, Centre Universitaire des Saints Peres, Université de Paris-Descartes , Paris , France
| |
Collapse
|
35
|
Regulatory T-cell vaccination independent of auto-antigen. Exp Mol Med 2014; 46:e82. [PMID: 24626168 PMCID: PMC3972794 DOI: 10.1038/emm.2014.4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/06/2013] [Indexed: 12/21/2022] Open
Abstract
To date, efforts to treat autoimmune diseases have primarily focused on the disease symptoms rather than on the cause of the disease. In large part, this is attributed to not knowing the responsible auto-antigens (auto-Ags) for driving the self-reactivity coupled with the poor success of treating autoimmune diseases using oral tolerance methods. Nonetheless, if tolerogenic approaches or methods that stimulate regulatory T (Treg) cells can be devised, these could subdue autoimmune diseases. To forward such efforts, our approach with colonization factor antigen I (CFA/I) fimbriae is to establish bystander immunity to ultimately drive the development of auto-Ag-specific Treg cells. Using an attenuated Salmonella vaccine expressing CFA/I fimbriae, fimbriae-specific Treg cells were induced without compromising the vaccine's capacity to protect against travelers' diarrhea or salmonellosis. By adapting the vaccine's anti-inflammatory properties, it was found that it could also dampen experimental inflammatory diseases resembling multiple sclerosis (MS) and rheumatoid arthritis. Because of this bystander effect, disease-specific Treg cells are eventually induced to resolve disease. Interestingly, this same vaccine could elicit the required Treg cell subset for each disease. For MS-like disease, conventional CD25+ Treg cells are stimulated, but for arthritis CD39+ Treg cells are induced instead. This review article will examine the potential of treating autoimmune diseases without having previous knowledge of the auto-Ag using an innocuous antigen to stimulate Treg cells via the production of transforming growth factor-β and interleukin-10.
Collapse
|
36
|
Lee J, Walsh MC, Hoehn KL, James DE, Wherry EJ, Choi Y. Regulator of fatty acid metabolism, acetyl coenzyme a carboxylase 1, controls T cell immunity. THE JOURNAL OF IMMUNOLOGY 2014; 192:3190-9. [PMID: 24567531 DOI: 10.4049/jimmunol.1302985] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Fatty acids (FAs) are essential constituents of cell membranes, signaling molecules, and bioenergetic substrates. Because CD8(+) T cells undergo both functional and metabolic changes during activation and differentiation, dynamic changes in FA metabolism also occur. However, the contributions of de novo lipogenesis to acquisition and maintenance of CD8(+) T cell function are unclear. In this article, we demonstrate the role of FA synthesis in CD8(+) T cell immunity. T cell-specific deletion of acetyl coenzyme A carboxylase 1 (ACC1), an enzyme that catalyzes conversion of acetyl coenzyme A to malonyl coenzyme A, a carbon donor for long-chain FA synthesis, resulted in impaired peripheral persistence and homeostatic proliferation of CD8(+) T cells in naive mice. Loss of ACC1 did not compromise effector CD8(+) T cell differentiation upon listeria infection but did result in a severe defect in Ag-specific CD8(+) T cell accumulation because of increased death of proliferating cells. Furthermore, in vitro mitogenic stimulation demonstrated that defective blasting and survival of ACC1-deficient CD8(+) T cells could be rescued by provision of exogenous FA. These results suggest an essential role for ACC1-mediated de novo lipogenesis as a regulator of CD8(+) T cell expansion, and may provide insights for therapeutic targets for interventions in autoimmune diseases, cancer, and chronic infections.
Collapse
Affiliation(s)
- JangEun Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | | | | | | | | | | |
Collapse
|
37
|
Clarkson BD, Héninger E, Harris MG, Lee J, Sandor M, Fabry Z. Innate-adaptive crosstalk: how dendritic cells shape immune responses in the CNS. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 946:309-33. [PMID: 21948376 DOI: 10.1007/978-1-4614-0106-3_18] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DCs) are a heterogeneous group of professional antigen presenting cells that lie in a nexus between innate and adaptive immunity because they recognize and respond to danger signals and subsequently initiate and regulate effector T-cell responses. Initially thought to be absent from the CNS, both plasmacytoid and conventional DCs as well as DC precursors have recently been detected in several CNS compartments where they are seemingly poised for responding to injury and pathogens. Additionally, monocyte-derived DCs rapidly accumulate in the inflamed CNS where they, along with other DC subsets, may function to locally regulate effector T-cells and/or carry antigens to CNS-draining cervical lymph nodes. In this review we highlight recent research showing that (a) distinct inflammatory stimuli differentially recruit DC subsets to the CNS; (b) DC recruitment across the blood-brain barrier (BBB) is regulated by adhesion molecules, growth factors, and chemokines; and (c) DCs positively or negatively regulate immune responses in the CNS.
Collapse
Affiliation(s)
- Benjamin D Clarkson
- Department of Pathology and Laboratory Medicine, 6130 MSC University of Wisconsin, School of Medicine and Public Health, Madison, WI 53706, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Nicolò C, Di Sante G, Migliara G, Valentini MG, Piermattei A, Delogu G, Ria F. Intracellular bacteria can cause EAE in SJL mice or modify self-specific T cell repertoire. J Neurol Sci 2011; 311:103-6. [PMID: 21917274 DOI: 10.1016/j.jns.2011.08.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 08/18/2011] [Accepted: 08/29/2011] [Indexed: 10/17/2022]
Abstract
Environment and genetic are both relevant in determining development of Multiple Sclerosis. Many epidemiological observations converge on indicating EBV infection and Vitamin D levels as major players among the environmental factors. Bacteria and bacterial products are however potent triggers of immune responses, and recent work from several laboratories indicates that the microbiota plays a prominent role in "priming" or protecting individuals for development of experimental autoimmune diseases. Here we report our recent work dealing with the role of non-pathogenic mycobacteria and their innate receptors in relapsing-remitting experimental autoimmune encephalomyelitis in the SJL mouse and in mobilization of CNS-reactive T cells. We finally discuss how bacteria are likely involved in the pathogenesis of Multiple Sclerosis, expecially with regard to their role in driving the recurring acute episodes of disease.
Collapse
Affiliation(s)
- Chiara Nicolò
- Institute of General Pathology, Università Cattolica del S. Cuore, L.go F.Vito, 1, 00168 Roma, Italy
| | | | | | | | | | | | | |
Collapse
|
39
|
Chatterjee S, Dwivedi VP, Singh Y, Siddiqui I, Sharma P, Van Kaer L, Chattopadhyay D, Das G. Early secreted antigen ESAT-6 of Mycobacterium tuberculosis promotes protective T helper 17 cell responses in a toll-like receptor-2-dependent manner. PLoS Pathog 2011; 7:e1002378. [PMID: 22102818 PMCID: PMC3213116 DOI: 10.1371/journal.ppat.1002378] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 09/29/2011] [Indexed: 12/22/2022] Open
Abstract
Despite its relatively poor efficacy, Bacillus Calmette-Guérin (BCG) has been used as a tuberculosis (TB) vaccine since its development in 1921. BCG induces robust T helper 1 (Th1) immune responses but, for many individuals, this is not sufficient for host resistance against Mycobacterium tuberculosis (M. tb) infection. Here we provide evidence that early secreted antigenic target protein 6 (ESAT-6), expressed by the virulent M. tb strain H37Rv but not by BCG, promotes vaccine-enhancing Th17 cell responses. These activities of ESAT-6 were dependent on TLR-2/MyD88 signalling and involved IL-6 and TGF-β production by dendritic cells. Thus, animals that were previously infected with H37Rv or recombinant BCG containing the RD1 region (BCG::RD1) exhibited improved protection upon re-challenge with virulent H37Rv compared with mice previously infected with BCG or RD1-deficient H37Rv (H37RvΔRD1). However, TLR-2 knockout (TLR-2⁻/⁻) animals neither showed Th17 responses nor exhibited improved protection in response to immunization with H37Rv. Furthermore, H37Rv and BCG::RD1 infection had little effect on the expression of the anti-inflammatory microRNA-146a (miR146a) in dendritic cells (DCs), whereas BCG and H37RvΔRD1 profoundly induced its expression in DCs. Consistent with these findings, ESAT-6 had no effect on miR146a expression in uninfected DCs, but dramatically inhibited its upregulation in BCG-infected or LPS-treated DCs. Collectively, our findings indicate that, in addition to Th1 immunity induced by BCG, RD1/ESAT-6-induced Th17 immune responses are essential for optimal vaccine efficacy.
Collapse
Affiliation(s)
- Samit Chatterjee
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Ved Prakash Dwivedi
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Yogesh Singh
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Imran Siddiqui
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Pawan Sharma
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Luc Van Kaer
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | | | - Gobardhan Das
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| |
Collapse
|
40
|
Cobb D, Hambright D, Smeltz RB. T-bet-independent effects of IL-12 family cytokines on regulation of Th17 responses to experimental T. cruzi infection. J Leukoc Biol 2010; 88:965-71. [PMID: 20807701 DOI: 10.1189/jlb.0410238] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Tbx21 (i.e., T-bet) is an IFN-γ-inducible transcription factor that promotes Th1 differentiation. Previously, we reported that Tbx21(-/-) mice develop a robust Th17 response to the parasite Trypanosoma cruzi, including CD4(+) T cell subsets producing IL-17 and IFN-γ. Because of the known inhibitory effects of IFN-γ on Th17 cells, the purpose of this study was to determine the contribution of IFN-γ to regulation of Th17 differentiation during the course of T. cruzi infection. We observed that infection of IFN-γ(-/-) or Stat-1(-/-) mice generated increased numbers of IL-17-producing cells. In sharp contrast to infected Stat-1(-/-) or Tbx21(-/-) mice, however, IFN-γ(-/-) mice developed a lower overall Th17 response, suggesting that IFN-γ was not required for T-bet-dependent activity, including T-bet-dependent expression of CXCR3. To determine if IFN-γ could influence Th17 responses indirectly by acting on APCs, we neutralized IFN-γ in cultures containing APC and T. cruzi antigens. Although anti-IFN-γ increased IL-17 production modestly, anti-IFN-γ and anti-IL-12 led to a significant enhancement of T. cruzi-specific IL-17 (P<0.01). In contrast to the inhibitory effects of IL-12, IL-23 was able to stimulate Tbx21(-/-) T cells and cause a striking increase in T. cruzi-specific IL-17. These data show that the IL-12 family of cytokines can influence Th17 responses in a T-bet-independent manner and that the effects of IFN-γ are not necessarily related to its ability to induce T-bet expression in T cells.
Collapse
Affiliation(s)
- Dustin Cobb
- Virginia Commonwealth University, Medical College of Virginia, Richmond, VA 23298, USA
| | | | | |
Collapse
|
41
|
Lee J, Sandor M, Heninger E, Fabry Z. Mycobacteria-induced suppression of autoimmunity in the central nervous system. J Neuroimmune Pharmacol 2010; 5:210-9. [PMID: 20333556 PMCID: PMC2875250 DOI: 10.1007/s11481-010-9199-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Accepted: 02/19/2010] [Indexed: 10/19/2022]
Abstract
Mycobacterial suppression of central nervous system (CNS) autoimmunity has been demonstrated in various experimental models, epidemiological studies, and clinical trials. Recent studies have led to an increased understanding of the cellular and molecular interactions involved in the pathogenesis of autoimmune diseases and of mycobacterial immunity. Here, we review some of the mechanisms by which mycobacterial infection might modulate the clinical course of CNS autoimmunity. A more complete understanding of these mechanisms may lead to the development of novel immunotherapeutic tools for treating autoimmune diseases.
Collapse
Affiliation(s)
- JangEun Lee
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA. Cellular and Molecular Pathology Program, School of Medicine and Public Health, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA. Cellular and Molecular Pathology Program, School of Medicine and Public Health, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| | - Erika Heninger
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA. Cellular and Molecular Pathology Program, School of Medicine and Public Health, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW It has often been suggested that autoimmune diseases are initiated by certain infectious agents that mimic self-antigens or polyclonally activated autoreactive lymphocytes. An alternative, and not necessarily mutually exclusive, hypothesis that some infections might inhibit the onset of some autoimmune conditions has more recently been explored. In this review, the evidence suggesting that the current rise in the incidence of some autoimmune diseases is attributable to a decrease in the incidence of exposure to certain infections will be discussed. RECENT FINDINGS Studies using animal models have shown that some infectious agents or products derived from them have the potential to inhibit the onset of autoimmunity. These studies have led to the suggestion that human autoimmune or allergic diseases might be alleviated by the use of microbial products. There are some data that would support such an observation. SUMMARY The incidence of some autoimmune diseases has increased dramatically in recent years in the developed world. Many autoimmune diseases are governed by both genetic and environmental factors. Our immune system has coevolved with infectious agents. There have been marked changes in the exposure to certain infectious agents over the last 70 years. It has been proposed that certain infections of historical importance might inhibit the development of autoimmune disorder. This review highlights studies addressing the ways in which infectious agents might inhibit onset of autoimmunity, and how this might lead to the development of novel therapeutic approaches.
Collapse
|
43
|
Lee J, Ling C, Kosmalski MM, Hulseberg P, Schreiber HA, Sandor M, Fabry Z. Intracerebral Mycobacterium bovis bacilli Calmette-Guerin infection-induced immune responses in the CNS. J Neuroimmunol 2009; 213:112-22. [PMID: 19535154 PMCID: PMC2937834 DOI: 10.1016/j.jneuroim.2009.05.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 05/18/2009] [Accepted: 05/18/2009] [Indexed: 12/17/2022]
Abstract
To study whether cerebral mycobacterial infection induces granuloma and protective immunity similar to systemic infection, we intracerebrally infected mice with Mycobacterium bovis bacilli Calmette-Guerin. Granuloma and IFN-gamma(+)CD4(+) T cell responses are induced in the central nervous system (CNS) similar to periphery, but the presence of IFN-gammaIL-17 double-positive CD4(+) T cells is unique to the CNS. The major CNS source of TNF-alpha is microglia, with modest production by CD4(+) T cells and macrophage. Protective immunity is accompanied by accumulation of Foxp3(+)CD4(+) T cells and PD-L2(+) dendritic cells, suggesting that both inflammatory and anti-inflammatory responses develop in the CNS following mycobacterial infection.
Collapse
Affiliation(s)
- JangEun Lee
- Department of Pathology and Laboratory Medicine, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
- Cellular and Molecular Pathology Program, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
| | - Changying Ling
- Department of Pathology and Laboratory Medicine, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
| | - Michelle M. Kosmalski
- Department of Pathology and Laboratory Medicine, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
| | - Paul Hulseberg
- Department of Pathology and Laboratory Medicine, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
- Cellular and Molecular Pathology Program, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
| | - Heidi A. Schreiber
- Department of Pathology and Laboratory Medicine, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
- Cellular and Molecular Pathology Program, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
- Cellular and Molecular Pathology Program, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
- Cellular and Molecular Pathology Program, University of Wisconsin, School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA
| |
Collapse
|
44
|
Guo S, Cobb D, Smeltz RB. T-bet inhibits the in vivo differentiation of parasite-specific CD4+ Th17 cells in a T cell-intrinsic manner. THE JOURNAL OF IMMUNOLOGY 2009; 182:6179-86. [PMID: 19414771 DOI: 10.4049/jimmunol.0803821] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD4(+) Th17 cells have emerged as a new T cell subset in the Th1/Th2 paradigm, and efforts have shifted toward understanding the factors that regulate their development in vivo. To analyze the role of the transcription factor T-bet in regulation of Th17 cells, we used a murine model of Trypanosoma cruzi infection, a protozoan parasite that causes Chagas disease in humans. Infection of Tbx21(-/-) mice led to normal, unimpaired development of Ag-specific CD4(+) T cells producing IFN-gamma. However, a robust Th17 response developed concomitant with Th1 responses. Despite significant IFN-gamma production, the physiological effects of Th17 responses prevailed as there was a sharp increase in Gr-1(+)Ly6G(+) neutrophils. Adoptive transfer of T cells from infected Tbx21(-/-) mice into Rag-2(-/-) mice (Tbx21(+/+)) revealed that CD4(+) T cells maintained their IL-17-producing phenotype, including those cells capable of producing both IFN-gamma and IL-17. Furthermore, and in contrast to the effects of IL-2 on Th17 development, IL-2 had no effect on IL-17 production by primed T cells. Importantly, adoptive transfer of T cells from naive Tbx21(-/-) mice into infected Rag-2(-/-) mice recapitulated the differentiation of T. cruzi-specific Th17 cells observed in infected Tbx21(-/-) mice. Conversely, transfer of wild-type T cells into infected Tbx21(-/-) mice did not reveal an increase in Th17 development. These results demonstrate that T-bet regulates the differentiation of T. cruzi-specific Th17 cells in vivo in a T cell-intrinsic manner. These data provide important insight into the role of T-bet in regulation of parasite-specific Th17 responses.
Collapse
Affiliation(s)
- Siqi Guo
- Department of Microbiology and Immunology, Virginia Commonwealth University, Medical College of Virginia Campus, Richmond, VA 23298, USA
| | | | | |
Collapse
|