1
|
Tian N, Chu H, Li Q, Sun H, Zhang J, Chu N, Sun Z. Host-directed therapy for tuberculosis. Eur J Med Res 2025; 30:267. [PMID: 40211397 PMCID: PMC11987284 DOI: 10.1186/s40001-025-02443-4] [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: 09/30/2024] [Accepted: 03/09/2025] [Indexed: 04/13/2025] Open
Abstract
Current TB treatment regimens are hindered by drug resistance, numerous adverse effects, and long treatment durations, highlighting the need for 'me-better' treatment regimens. Host-directed therapy (HDT) has gained recognition as a promising approach in TB treatment. It allows the repurposing of existing drugs approved for other conditions and aims to enhance the effectiveness of existing anti-TB therapies, minimize drug resistance, decrease treatment duration, and adverse effects. By modulating the host immune response, HDT ameliorates immunopathological damage and improves overall outcomes by promoting autophagy, antimicrobial peptide production, and other mechanisms. It holds promise for addressing the challenges posed by multiple and extensively drug-resistant Mycobacterium tuberculosis strains, which are increasingly difficult to treat using conventional therapies. This article reviews various HDT candidates, including repurposed drugs, explores their underlying mechanisms such as autophagy promotion and inflammation reduction, while emphasizing their potential to improve TB treatment outcomes and outlining future research directions.
Collapse
Affiliation(s)
- Na Tian
- Department of Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Qi Li
- Department of Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Hong Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Jingfang Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Naihui Chu
- Department of Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
2
|
Kalsum S, Xu R, Akber M, Huang S, Lerm M, Chen Y, Lourda M, Zhou Y, Brighenti S. Dual GSK-3β/HDAC Inhibitors Enhance the Efficacy of Macrophages to Control Mycobacterium tuberculosis Infection. Biomolecules 2025; 15:550. [PMID: 40305296 PMCID: PMC12024928 DOI: 10.3390/biom15040550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/29/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Multitarget drug discovery, including host-directed therapy, is particularly promising for tuberculosis (TB) due to the resilience of Mycobacterium tuberculosis (Mtb) as well as the complexity of the host's immune response. In this proof-of-concept study, we used high-content imaging to test a novel panel of dual glycogen synthase kinase 3 beta (GSK-3β) and histone deacetylase (HDAC) 1 and 6 inhibitor candidates for their efficacy in reducing the growth of green fluorescent protein (GFP)-expressing mycobacteria in human primary macrophages. We demonstrate that all ten test compounds, also including the GSK-3β inhibitor SB415286, exhibit an antimycobacterial effect of 20-60% at low micromolar doses and are non-toxic to host cells. Mtb growth showed a positive correlation with the respective 50% inhibitory concentration (IC50) values of GSK-3β, HDAC1, and HDAC6 in each compound, indicating that compounds with a potent IC50 value for HDAC1, in particular, corresponded to higher antimycobacterial activity. Furthermore, the results from multiparametric flow cytometry and a customized multiplex RNA array demonstrated that SB415286 and selected compounds, C02 and C06, could modulate immune polarization and inflammation in Mtb-infected macrophages involving an enhanced expression of CCL2, IL-10 and S100A9, but a decrease in inflammatory mediators including COX-2, TNF-α, and NFκB. These data suggest that GSK-3β inhibition alone can decrease the intracellular growth of mycobacteria and regulate macrophage inflammation, while dual GSK-3β/HDAC inhibitors enhance this efficacy. Accordingly, the tailored design of dual GSK-3β/HDAC inhibitors could represent an innovative approach to host-directed therapy in TB.
Collapse
Affiliation(s)
- Sadaf Kalsum
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, 141 52 Huddinge, Sweden; (S.K.); (R.X.); (M.A.)
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden;
| | - Ruilan Xu
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, 141 52 Huddinge, Sweden; (S.K.); (R.X.); (M.A.)
| | - Mira Akber
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, 141 52 Huddinge, Sweden; (S.K.); (R.X.); (M.A.)
| | - Shengjie Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China; (S.H.); (Y.C.)
| | - Maria Lerm
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden;
| | - Yuqing Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China; (S.H.); (Y.C.)
| | - Magda Lourda
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, 141 52 Huddinge, Sweden;
| | - Yang Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China; (S.H.); (Y.C.)
| | - Susanna Brighenti
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, 141 52 Huddinge, Sweden; (S.K.); (R.X.); (M.A.)
| |
Collapse
|
3
|
Kalsum S, Akber M, Loreti MG, Andersson B, Danielson E, Lerm M, Brighenti S. Sirtuin inhibitors reduce intracellular growth of M. tuberculosis in human macrophages via modulation of host cell immunity. Sci Rep 2024; 14:28150. [PMID: 39548210 PMCID: PMC11568201 DOI: 10.1038/s41598-024-79136-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Host-directed therapies aiming to strengthen the body's immune system, represent an underexplored opportunity to improve treatment of tuberculosis (TB). We have previously shown in Mycobacterium tuberculosis (Mtb)-infection models and clinical trials that treatment with the histone deacetylase (HDAC) inhibitor, phenylbutyrate (PBA), can restore Mtb-induced impairment of antimicrobial responses and improve clinical outcomes in pulmonary TB. In this study, we evaluated the efficacy of different groups of HDAC inhibitors to reduce Mtb growth in human immune cells. A panel of 21 selected HDAC inhibitors with different specificities that are known to modulate infection or inflammation was tested using high-content live-cell imaging and analysis. Monocyte-derived macrophages or bulk peripheral blood cells (PBMCs) were infected with the green fluorescent protein (GFP)-expressing Mtb strains H37Ra or H37Rv and treated with HDAC inhibitors in the micromolar range in parallel with a combination of the first-line antibiotics, rifampicin, and isoniazid. Host cell viability in HDAC inhibitor treated cell cultures was monitored with Cytotox-red. Seven HDAC inhibitors were identified that reduced Mtb growth in macrophages > 45-75% compared to average 40% for PBA. The most effective compounds were inhibitors of the class III HDAC proteins, the sirtuins. While these compounds may exhibit their effects by improving macrophage function, one of the sirtuin inhibitors, tenovin, was also highly effective in extracellular killing of Mtb bacilli. Antimicrobial synergy testing using checkerboard assays revealed additive effects between selected sirtuin inhibitors and subinhibitory concentrations of rifampicin or isoniazid. A customized macrophage RNA array including 23 genes associated with cytokines, chemokines and inflammation, suggested that Mtb-infected macrophages are differentially modulated by the sirtuin inhibitors as compared to PBA. Altogether, these results demonstrated that sirtuin inhibitors may be further explored as promising host-directed compounds to support immune functions and reduce intracellular growth of Mtb in human cells.
Collapse
Affiliation(s)
- Sadaf Kalsum
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, Huddinge, 141 52, Sweden
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, 581 83, Sweden
| | - Mira Akber
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, Huddinge, 141 52, Sweden
| | - Marco Giulio Loreti
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, Huddinge, 141 52, Sweden
| | - Blanka Andersson
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, 581 83, Sweden
| | - Eva Danielson
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, 581 83, Sweden
| | - Maria Lerm
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, 581 83, Sweden
| | - Susanna Brighenti
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, Huddinge, 141 52, Sweden.
| |
Collapse
|
4
|
Whitmore M, Tobin I, Burkardt A, Zhang G. Nutritional Modulation of Host Defense Peptide Synthesis: A Novel Host-Directed Antimicrobial Therapeutic Strategy? Adv Nutr 2024; 15:100277. [PMID: 39053604 PMCID: PMC11381887 DOI: 10.1016/j.advnut.2024.100277] [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: 02/28/2024] [Revised: 06/11/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
The escalating threat of antimicrobial resistance underscores the imperative for innovative therapeutic strategies. Host defense peptides (HDPs), integral components of innate immunity, exhibit profound antimicrobial and immunomodulatory properties. Various dietary compounds, such as short-chain fatty acids, vitamins, minerals, sugars, amino acids, phytochemicals, bile acids, probiotics, and prebiotics have been identified to enhance the synthesis of endogenous HDPs without provoking inflammatory response or compromising barrier integrity. Additionally, different classes of these compounds synergize in augmenting HDP synthesis and disease resistance. Moreover, dietary supplementation of several HDP-inducing compounds or their combinations have demonstrated robust protection in rodents, rabbits, pigs, cattle, and chickens from experimental infections. However, the efficacy of these compounds in inducing HDP synthesis varies considerably among distinct compounds. Additionally, the regulation of HDP genes occurs in a gene-specific, cell type-specific, and species-specific manner. In this comprehensive review, we systematically summarized the modulation of HDP synthesis and the mechanism of action attributed to each major class of dietary compounds, including their synergistic combinations, across a spectrum of animal species including humans. We argue that the ability to enhance innate immunity and barrier function without triggering inflammation or microbial resistance positions the nutritional modulation of endogenous HDP synthesis as a promising host-directed approach for mitigating infectious diseases and antimicrobial resistance. These HDP-inducing compounds, particularly in combinations, harbor substantial clinical potential for further exploration in antimicrobial therapies for both human and other animals.
Collapse
Affiliation(s)
- Melanie Whitmore
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Isabel Tobin
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Amanda Burkardt
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States.
| |
Collapse
|
5
|
Helaine S, Conlon BP, Davis KM, Russell DG. Host stress drives tolerance and persistence: The bane of anti-microbial therapeutics. Cell Host Microbe 2024; 32:852-862. [PMID: 38870901 PMCID: PMC11446042 DOI: 10.1016/j.chom.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/03/2024] [Accepted: 04/25/2024] [Indexed: 06/15/2024]
Abstract
Antibiotic resistance, typically associated with genetic changes within a bacterial population, is a frequent contributor to antibiotic treatment failures. Antibiotic persistence and tolerance, which we collectively term recalcitrance, represent transient phenotypic changes in the bacterial population that prolong survival in the presence of typically lethal concentrations of antibiotics. Antibiotic recalcitrance is challenging to detect and investigate-traditionally studied under in vitro conditions, our understanding during infection and its contribution to antibiotic failure is limited. Recently, significant progress has been made in the study of antibiotic-recalcitrant populations in pathogenic species, including Mycobacterium tuberculosis, Staphylococcus aureus, Salmonella enterica, and Yersiniae, in the context of the host environment. Despite the diversity of these pathogens and infection models, shared signals and responses promote recalcitrance, and common features and vulnerabilities of persisters and tolerant bacteria have emerged. These will be discussed here, along with progress toward developing therapeutic interventions to better treat recalcitrant pathogens.
Collapse
Affiliation(s)
- Sophie Helaine
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
| | - Brian P Conlon
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| | - Kimberly M Davis
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
6
|
Shapira T, Christofferson M, Av-Gay Y. The antimicrobial activity of innate host-directed therapies: A systematic review. Int J Antimicrob Agents 2024; 63:107138. [PMID: 38490573 DOI: 10.1016/j.ijantimicag.2024.107138] [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: 10/01/2023] [Revised: 02/23/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024]
Abstract
Intracellular human pathogens are the deadliest infectious diseases and are difficult to treat effectively due to their protection inside the host cell and the development of antimicrobial resistance (AMR). An emerging approach to combat these intracellular pathogens is host-directed therapies (HDT), which harness the innate immunity of host cells. HDT rely on small molecules to promote host protection mechanisms that ultimately lead to pathogen clearance. These therapies are hypothesized to: (1) possess indirect yet broad, cross-species antimicrobial activity, (2) effectively target drug-resistant pathogens, (3) carry a reduced susceptibility to the development of AMR and (4) have synergistic action with conventional antimicrobials. As the field of HDT expands, this systematic review was conducted to collect a compendium of HDT and their characteristics, such as the host mechanisms affected, the pathogen inhibited, the concentrations investigated and the magnitude of pathogen inhibition. The evidential support for the main four HDT hypotheses was assessed and concluded that HDT demonstrate robust cross-species activity, are active against AMR pathogens, clinical isolates and laboratory-adapted pathogens. However, limited information exists to support the notion that HDT are synergistic with canonical antimicrobials and are less predisposed to AMR development.
Collapse
Affiliation(s)
- Tirosh Shapira
- Department of Medicine, Division of Infectious Disease, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Matthew Christofferson
- Department of Microbiology and Immunology, Division of Infectious Disease, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Yossef Av-Gay
- Department of Medicine, Division of Infectious Disease, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada; Department of Microbiology and Immunology, Division of Infectious Disease, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
7
|
Chandra H, Rahman A, Yadav P, Maurya G, Kumar Shukla S. Effect of adjunct Vitamin D treatment in vitamin D deficient pulmonary tuberculosis patients: A randomized, double blind, active controlled clinical trial. Indian J Tuberc 2024; 71:170-178. [PMID: 38589121 DOI: 10.1016/j.ijtb.2023.04.026] [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: 03/18/2023] [Revised: 04/15/2023] [Accepted: 04/28/2023] [Indexed: 04/10/2024]
Abstract
BACKGROUND Since, Vitamin D [1α,25(OH)2D)] enhances antimicrobial activity of Innate immunity and modulate Adaptive immune responses, simultaneously, so it play a potential role for balanced immune activity against Mycobacterium tuberculosis and restricting tissue injuries within the TB patients.(Chun et al., 2011) 9 We aimed to determine the role of adjunct Vitamin D treatment on the outcome of pulmonary tuberculosis patients and evaluated the effect of Vitamin D administration on Differential Leucocyte Count, Erythrocyte Sedimentation Rate, serum Adenosine deaminase, serum C- reactive protein, Oxygen saturation (SpO2) and Body Weight in Vitamin D deficient pulmonary tuberculosis patients. METHODS We conducted a prospective, interventional, randomized, double blind, parallel group, active controlled clinical trial. Newly diagnosed Vitamin D deficient pulmonary tuberculosis patients were randomly assigned to intervention group (received standard anti-tubercular treatment with adjunct Vitamin D3) and control group (received standard anti-tubercular treatment without adjunct Vitamin D3). Total four doses [each dose of 2.5 mg (100000 IU)] of Vitamin D3 were given, orally. First dose was given within 7 days of starting anti-tubercular treatment and second, third, fourth dose were given at 2, 4 and 6 weeks respectively. At the time of enrollment, we measured all baseline characteristics. During follow-up, we measured the study variables and monitored adverse events at 2, 4, 6, 8 and 12 weeks. Our safety parameter was serum corrected calcium level to assess the risk of hypercalcemia. RESULTS Total 130 pulmonary TB patients, 65 patients in each group, were analyzed. Our study results showed that decrease in Neutrophil count was statistically significant with small effect sizes at every time point of measurement and increase in Lymphocyte count was statistically significant with small and moderate effect sizes at 4, 6 and 8 week for intervention group than for control group. Decrease in erythrocyte sedimentation rate was statistically significant with small effect sizes at 6 and 8 week, decrease in serum adenosine deaminase and serum C- reactive protein was statistically significant with moderate effect sizes at 4, 6 and 8 week for intervention group than for control group. Increase in Oxygen saturation was statistically significant at 4 week with small effect size and increase in body weight was statistically significant with small effect sizes for intervention group than for control group. No case of hypercalcemia was reported. CONCLUSION Our findings suggest a potential role of adjunctive Vitamin D3 to accelerate resolution of inflammatory responses and improvement in clinical outcomes of pulmonary TB patients. TRIAL REGISTRATION This trial is registered with Clinical Trials Registry - INDIA (http://ctri.nic.in) with CTRI Number - CTRI/2021/11/037914. PLACE OF STUDY Room Number 27, first floor out-patients department (OPD) and inpatient Wards, fourth floor, Department of Respiratory Medicine, Uttar Pradesh University of Medical Sciences, Saifai, Etawah (U.P.), INDIA.
Collapse
Affiliation(s)
- Harish Chandra
- Department of Biochemistry, Academic Block, Uttar Pradesh University of Medical Sciences, Saifai, Etawah, (U.P), 206130, India.
| | - Adil Rahman
- Department of Biochemistry, Academic Block, Uttar Pradesh University of Medical Sciences, Saifai, Etawah, (U.P), 206130, India
| | - Prashant Yadav
- Department of Respiratory Medicine, Uttar Pradesh University of Medical Sciences, Saifai, Etawah, (U.P.), 206130, India
| | - Geeta Maurya
- Department of Pathology, Academic Block, Uttar Pradesh University of Medical Sciences, Saifai, Etawah, (U.P.), 206130, India
| | - Sushil Kumar Shukla
- Department of Community Medicine, Academic Block, Uttar Pradesh University of Medical Sciences, Saifai, Etawah, (U.P.), 206130, India
| |
Collapse
|
8
|
Lim S, Lee D, Jeong S, Park JW, Im J, Choi B, Gwak D, Yun CH, Seo HS, Han SH. Serotype-Dependent Inhibition of Streptococcus pneumoniae Growth by Short-Chain Fatty Acids. J Microbiol Biotechnol 2024; 34:47-55. [PMID: 38044707 PMCID: PMC10840490 DOI: 10.4014/jmb.2309.09003] [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: 09/04/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 12/05/2023]
Abstract
Streptococcus pneumoniae (pneumococcus) is an opportunistic pathogen that can cause severe infectious diseases such as pneumonia, meningitis, and otitis media. Despite the availability of antibiotics and pneumococcal vaccines against some invasive serotypes, pneumococcal infection remains a tremendous clinical challenge due to the increasing frequency of infection by antimicrobial resistant, nonencapsulated, and/or non-vaccine serotype strains. Short-chain fatty acids (SCFAs), which are produced at various mucosal sites in the body, have potent antimicrobial activity, including inhibition of pathogen growth and/or bacterial biofilm formation. In this study, we investigated the antimicrobial activity of SCFAs (acetate, propionate, and butyrate) against various serotypes pneumococci. Propionate generally inhibited the growth of S. pneumoniae serotypes included in the pneumococcal conjugate vaccine (PCV) 13, except for serotypes 3 and 7F, though butyrate and acetate showed no or low inhibition, depending on the serotypes. Of note, butyrate showed strong inhibition against serotype 3, the most prevalent invasive strain since the introduction of the PCV. No SCFAs showed inhibitory effects against serotype 7F. Remarkably, the nonencapsulated pneumococcal strain had more sensitivity to SCFAs than encapsulated parental strains. Taken together, these results suggest that propionate showing the most potent inhibition of pneumococcal growth may be used as an alternative treatment for pneumococcal infection, and that butyrate could be used against serotype 3, which is becoming a serious threat.
Collapse
Affiliation(s)
- Suwon Lim
- Department of Oral Microbiology Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Dongwook Lee
- Department of Oral Microbiology Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Sungho Jeong
- Department of Oral Microbiology Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong Woo Park
- Department of Oral Microbiology Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Jintaek Im
- Department of Oral Microbiology Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Bokeum Choi
- Department of Oral Microbiology Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Donghyun Gwak
- Department of Oral Microbiology Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Institutes of Green Bio Science and Technology, Seoul National University, Pyeongchang 25354, Republic of Korea
| | - Ho Seong Seo
- Research Division for Biotechnology, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
9
|
Nguyen M, Ahn P, Dawi J, Gargaloyan A, Kiriaki A, Shou T, Wu K, Yazdan K, Venketaraman V. The Interplay between Mycobacterium tuberculosis and Human Microbiome. Clin Pract 2024; 14:198-213. [PMID: 38391403 PMCID: PMC10887847 DOI: 10.3390/clinpract14010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/24/2024] Open
Abstract
Tuberculosis (TB), a respiratory disease caused by Mycobacterium tuberculosis (Mtb), is a significant cause of mortality worldwide. The lung, a breeding ground for Mtb, was once thought to be a sterile environment, but has now been found to host its own profile of microbes. These microbes are critical in the development of the host immune system and can produce metabolites that aid in host defense against various pathogens. Mtb infection as well as antibiotics can shift the microbial profile, causing dysbiosis and dampening the host immune response. Additionally, increasing cases of drug resistant TB have impacted the success rates of the traditional therapies of isoniazid, rifampin, pyrazinamide, and ethambutol. Recent years have produced tremendous research into the human microbiome and its role in contributing to or attenuating disease processes. Potential treatments aimed at altering the gut-lung bacterial axis may offer promising results against drug resistant TB and help mitigate the effects of TB.
Collapse
Affiliation(s)
- Michelle Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Phillip Ahn
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - John Dawi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Areg Gargaloyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Anthony Kiriaki
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Tiffany Shou
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Kevin Wu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Kian Yazdan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
10
|
Rodríguez-Carlos A, Jacobo-Delgado Y, Santos-Mena AO, García-Hernández MH, De Jesus-Gonzalez LA, Lara-Ramirez EE, Rivas-Santiago B. Histone deacetylase (HDAC) inhibitors- based drugs are effective to control Mycobacterium tuberculosis infection and promote the sensibility for rifampicin in MDR strain. Mem Inst Oswaldo Cruz 2023; 118:e230143. [PMID: 38126492 PMCID: PMC10740574 DOI: 10.1590/0074-02760230143] [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: 08/15/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Tuberculosis (TB) is a major public health problem, which has been aggravated by the alarming growth of drug-resistant tuberculosis. Therefore, the development of a safer and more effective treatment is needed. OBJECTIVES The aim of this work was repositioning and evaluate histone deacetylases (HDAC) inhibitors- based drugs with potential antimycobacterial activity. METHODS Using an in silico pharmacological repositioning strategy, three molecules that bind to the catalytic site of histone deacetylase were selected. Pneumocytes type II and macrophages were infected with Mycobacterium tuberculosis and treated with pre-selected HDAC inhibitors (HDACi). Subsequently, the ability of each of these molecules to directly promote the elimination of M. tuberculosis was evaluated by colony-forming unit (CFU)/mL. We assessed the expression of antimicrobial peptides and respiratory burst using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). FINDINGS Aminoacetanilide (ACE), N-Boc-1,2-phenylenediamine (N-BOC), 1,3-Diphenylurea (DFU), reduce bacillary loads in macrophages and increase the production of β-defensin-2, LL-37, superoxide dismutase (SOD) 3 and inducible nitric oxide synthase (iNOS). While only the use of ACE in type II pneumocytes decreases the bacterial load through increasing LL-37 expression. Furthermore, the use of ACE and rifampicin inhibited the survival of intracellular multi-drug resistance M. tuberculosis. MAIN CONCLUSIONS Our data support the usefulness of in silico approaches for drug repositioning to provide a potential adjunctive therapy for TB.
Collapse
Affiliation(s)
- Adrián Rodríguez-Carlos
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| | - Yolanda Jacobo-Delgado
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| | | | | | | | - Edgar E Lara-Ramirez
- Instituto Politécnico Nacional, Centro de Biotecnología Genómica, Laboratorio de Biotecnología Farmacéutica, Reynosa, Mexico
| | - Bruno Rivas-Santiago
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| |
Collapse
|
11
|
Kroon EE, Correa-Macedo W, Evans R, Seeger A, Engelbrecht L, Kriel JA, Loos B, Okugbeni N, Orlova M, Cassart P, Kinnear CJ, Tromp GC, Möller M, Wilkinson RJ, Coussens AK, Schurr E, Hoal EG. Neutrophil extracellular trap formation and gene programs distinguish TST/IGRA sensitization outcomes among Mycobacterium tuberculosis exposed persons living with HIV. PLoS Genet 2023; 19:e1010888. [PMID: 37616312 PMCID: PMC10470897 DOI: 10.1371/journal.pgen.1010888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/31/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
Persons living with HIV (PLWH) have an increased risk for tuberculosis (TB). After prolonged and repeated exposure, some PLWH never develop TB and show no evidence of immune sensitization to Mycobacterium tuberculosis (Mtb) as defined by persistently negative tuberculin skin tests (TST) and interferon gamma release assays (IGRA). This group has been identified and defined as HIV+ persistently TB, tuberculin and IGRA negative (HITTIN). To investigate potential innate mechanisms unique to individuals with the HITTIN phenotype we compared their neutrophil Mtb infection response to that of PLWH, with no TB history, but who test persistently IGRA positive, and tuberculin positive (HIT). Neutrophil samples from 17 HITTIN (PMNHITTIN) and 11 HIT (PMNHIT) were isolated and infected with Mtb H37Rv for 1h and 6h. RNA was extracted and used for RNAseq analysis. Since there was no significant differential transcriptional response at 1h between infected PMNHITTIN and PMNHIT, we focused on the 6h timepoint. When compared to uninfected PMN, PMNHITTIN displayed 3106 significantly upregulated and 3548 significantly downregulated differentially expressed genes (DEGs) (absolute cutoff of a log2FC of 0.2, FDR < 0.05) whereas PMNHIT demonstrated 3816 significantly upregulated and 3794 significantly downregulated DEGs following 6h Mtb infection. Contrasting the log2FC 6h infection response to Mtb from PMNHITTIN against PMNHIT, 2285 genes showed significant differential response between the two groups. Overall PMNHITTIN had a lower fold change response to Mtb infection compared to PMNHIT. According to pathway enrichment, Apoptosis and NETosis were differentially regulated between HITTIN and HIT PMN responses after 6h Mtb infection. To corroborate the blunted NETosis transcriptional response measured among HITTIN, fluorescence microscopy revealed relatively lower neutrophil extracellular trap formation and cell loss in PMNHITTIN compared to PMNHIT, showing that PMNHITTIN have a distinct response to Mtb.
Collapse
Affiliation(s)
- Elouise E. Kroon
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Wilian Correa-Macedo
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill International TB Centre, McGill University, Montréal, Canada
- Department of Biochemistry, McGill University, Montréal, Canada
| | - Rachel Evans
- Infectious Diseases and Immune Defence Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department Medical Biology (WEHI), Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Allison Seeger
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Department of Medicine, University of Cape Town, Observatory, South Africa
| | - Lize Engelbrecht
- Central Analytical Facilities, Microscopy Unit, Stellenbosch University, Cape Town, South Africa
| | - Jurgen A. Kriel
- Central Analytical Facilities, Microscopy Unit, Stellenbosch University, Cape Town, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Naomi Okugbeni
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Genomics Platform, Tygerberg, South Africa
| | - Marianna Orlova
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill International TB Centre, McGill University, Montréal, Canada
- Department of Biochemistry, McGill University, Montréal, Canada
| | - Pauline Cassart
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill International TB Centre, McGill University, Montréal, Canada
| | - Craig J. Kinnear
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Genomics Platform, Tygerberg, South Africa
| | - Gerard C. Tromp
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Centre for Bioinformatics and Computational Biology, University of Stellenbosch, Cape Town, South Africa
- SAMRC-SHIP South African Tuberculosis Bioinformatics Initiative (SATBBI), Center for Bioinformatics and Computational Biology, Cape Town, South Africa
| | - Marlo Möller
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Centre for Bioinformatics and Computational Biology, University of Stellenbosch, Cape Town, South Africa
| | - Robert J. Wilkinson
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Department of Medicine, University of Cape Town, Observatory, South Africa
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | - Anna K. Coussens
- Infectious Diseases and Immune Defence Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department Medical Biology (WEHI), Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Department of Medicine, University of Cape Town, Observatory, South Africa
| | - Erwin Schurr
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill International TB Centre, McGill University, Montréal, Canada
- Department of Biochemistry, McGill University, Montréal, Canada
| | - Eileen G. Hoal
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
12
|
Abstract
Mycobacteria are responsible for several human and animal diseases. NOD2 is a pattern recognition receptor that has an important role in mycobacterial recognition. However, the mechanisms by which mutations in NOD2 alter the course of mycobacterial infection remain unclear. Herein, we aimed to review the totality of studies directly addressing the relationship between NOD2 and mycobacteria as a foundation for moving the field forward. NOD2 was linked to mycobacterial infection at 3 levels: (1) genetic, through association with mycobacterial diseases of humans; (2) chemical, through the distinct NOD2 ligand in the mycobacterial cell wall; and (3) immunologic, through heightened NOD2 signaling caused by the unique modification of the NOD2 ligand. The immune response to mycobacteria is shaped by NOD2 signaling, responsible for NF-κB and MAPK activation, and the production of various immune effectors like cytokines and nitric oxide, with some evidence linking this to bacteriologic control. Absence of NOD2 during mycobacterial infection of mice can be detrimental, but the mechanism remains unknown. Conversely, the success of immunization with mycobacteria has been linked to NOD2 signaling and NOD2 has been targeted as an avenue of immunotherapy for diseases even beyond mycobacteria. The mycobacteria-NOD2 interaction remains an important area of study, which may shed light on immune mechanisms in disease.
Collapse
Affiliation(s)
- Jean-Yves Dubé
- Department of Microbiology and Immunology, McGill University, Montréal, Canada
| | - Marcel A. Behr
- Department of Medicine, McGill University Health Centre, Montréal, Canada
| |
Collapse
|
13
|
Yu Z, Shen X, Wang A, Hu C, Chen J. The gut microbiome: A line of defense against tuberculosis development. Front Cell Infect Microbiol 2023; 13:1149679. [PMID: 37143744 PMCID: PMC10152471 DOI: 10.3389/fcimb.2023.1149679] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 03/29/2023] [Indexed: 05/06/2023] Open
Abstract
The tuberculosis (TB) burden remains a significant global public health concern, especially in less developed countries. While pulmonary tuberculosis (PTB) is the most common form of the disease, extrapulmonary tuberculosis, particularly intestinal TB (ITB), which is mostly secondary to PTB, is also a significant issue. With the development of sequencing technologies, recent studies have investigated the potential role of the gut microbiome in TB development. In this review, we summarized studies investigating the gut microbiome in both PTB and ITB patients (secondary to PTB) compared with healthy controls. Both PTB and ITB patients show reduced gut microbiome diversity characterized by reduced Firmicutes and elevated opportunistic pathogens colonization; Bacteroides and Prevotella were reported with opposite alteration in PTB and ITB patients. The alteration reported in TB patients may lead to a disequilibrium in metabolites such as short-chain fatty acid (SCFA) production, which may recast the lung microbiome and immunity via the "gut-lung axis". These findings may also shed light on the colonization of Mycobacterium tuberculosis in the gastrointestinal tract and the development of ITB in PTB patients. The findings highlight the crucial role of the gut microbiome in TB, particularly in ITB development, and suggest that probiotics and postbiotics might be useful supplements in shaping a balanced gut microbiome during TB treatment.
Collapse
Affiliation(s)
- Ziqi Yu
- Munich Medical Research School, Ludwig Maximilian University of Munich (LMU), Munich, Germany
| | - Xiang Shen
- Munich Medical Research School, Ludwig Maximilian University of Munich (LMU), Munich, Germany
| | - Aiyao Wang
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People’s Hospital, Nanchang, Jiangxi, China
| | - Chong Hu
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People’s Hospital, Nanchang, Jiangxi, China
| | - Jianyong Chen
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People’s Hospital, Nanchang, Jiangxi, China
- *Correspondence: Jianyong Chen,
| |
Collapse
|
14
|
Inflammation-mediated tissue damage in pulmonary tuberculosis and host-directed therapeutic strategies. Semin Immunol 2023; 65:101672. [PMID: 36469987 DOI: 10.1016/j.smim.2022.101672] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 12/04/2022]
Abstract
Treatment of tuberculosis (TB) involves the administration of anti-mycobacterial drugs for several months. The emergence of drug-resistant strains of Mycobacterium tuberculosis (Mtb, the causative agent) together with increased disease severity in people with co-morbidities such as diabetes mellitus and HIV have hampered efforts to reduce case fatality. In severe disease, TB pathology is largely attributable to over-exuberant host immune responses targeted at controlling bacterial replication. Non-resolving inflammation driven by host pro-inflammatory mediators in response to high bacterial load leads to pulmonary pathology including cavitation and fibrosis. The need to improve clinical outcomes and reduce treatment times has led to a two-pronged approach involving the development of novel antimicrobials as well as host-directed therapies (HDT) that favourably modulate immune responses to Mtb. HDT strategies incorporate aspects of immune modulation aimed at downregulating non-productive inflammatory responses and augmenting antimicrobial effector mechanisms to minimise pulmonary pathology and accelerate symptom resolution. HDT in combination with existing antimycobacterial agents offers a potentially promising strategy to improve the long-term outcome for TB patients. In this review, we describe components of the host immune response that contribute to inflammation and tissue damage in pulmonary TB, including cytokines, matrix metalloproteinases, lipid mediators, and neutrophil extracellular traps. We then proceed to review HDT directed at these pathways.
Collapse
|
15
|
Secretory proteins of
Mycobacterium tuberculosis
and their roles in modulation of host immune responses: focus on therapeutic targets. FEBS J 2022; 289:4146-4171. [DOI: 10.1111/febs.16369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 01/04/2022] [Accepted: 01/21/2022] [Indexed: 12/01/2022]
|
16
|
Pattanaik KP, Sengupta S, Jit BP, Kotak R, Sonawane A. Host-Mycobacteria conflict: Immune responses of the host vs. the mycobacteria TLR2 and TLR4 ligands and concomitant host-directed therapy. Microbiol Res 2022; 264:127153. [DOI: 10.1016/j.micres.2022.127153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/21/2022] [Accepted: 07/29/2022] [Indexed: 12/15/2022]
|
17
|
Khoza LJ, Kumar P, Dube A, Demana PH, Choonara YE. Insights into Innovative Therapeutics for Drug-Resistant Tuberculosis: Host-Directed Therapy and Autophagy Inducing Modified Nanoparticles. Int J Pharm 2022; 622:121893. [PMID: 35680110 PMCID: PMC9169426 DOI: 10.1016/j.ijpharm.2022.121893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 10/25/2022]
|
18
|
Expression of Vitamin D Receptor (VDR) gene and VDR polymorphism rs11574113 in pulmonary tuberculosis patients and their household contacts. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Yang Q, Whitmore MA, Robinson K, Lyu W, Zhang G. Butyrate, Forskolin, and Lactose Synergistically Enhance Disease Resistance by Inducing the Expression of the Genes Involved in Innate Host Defense and Barrier Function. Antibiotics (Basel) 2021; 10:antibiotics10101175. [PMID: 34680756 PMCID: PMC8532606 DOI: 10.3390/antibiotics10101175] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 01/28/2023] Open
Abstract
The rising concern of antimicrobial resistance highlights a need for effective alternatives to antibiotics for livestock production. Butyrate, forskolin, and lactose are three natural products known to induce the synthesis of host defense peptides (HDP), which are a critical component of innate immunity. In this study, the synergy among butyrate, forskolin, and lactose in enhancing innate host defense, barrier function, and resistance to necrotic enteritis and coccidiosis was investigated. Our results indicated that the three compounds synergistically augmented the expressions of multiple HDP and barrier function genes in chicken HD11 macrophages. The compounds also showed an obvious synergy in promoting HDP gene expressions in chicken jejunal explants. Dietary supplementation of a combination of 1 g/kg sodium butyrate, 10 mg/kg forskolin-containing plant extract, and 10 g/kg lactose dramatically improved the survival of chickens from 39% to 94% (p < 0.001) in a co-infection model of necrotic enteritis. Furthermore, the three compounds largely reversed growth suppression, significantly alleviated intestinal lesions, and reduced colonization of Clostridium perfringens or Eimeria maxima in chickens with necrotic enteritis and coccidiosis (p < 0.01). Collectively, dietary supplementation of butyrate, forskolin, and lactose is a promising antibiotic alternative approach to disease control and prevention for poultry and possibly other livestock species.
Collapse
Affiliation(s)
- Qing Yang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (Q.Y.); (M.A.W.); (K.R.); (W.L.)
| | - Melanie A. Whitmore
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (Q.Y.); (M.A.W.); (K.R.); (W.L.)
| | - Kelsy Robinson
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (Q.Y.); (M.A.W.); (K.R.); (W.L.)
- Poultry Production and Product Safety Research Unit, USDA–Agricultural Research Service, Fayetteville, AR 72701, USA
| | - Wentao Lyu
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (Q.Y.); (M.A.W.); (K.R.); (W.L.)
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (Q.Y.); (M.A.W.); (K.R.); (W.L.)
- Correspondence: ; Tel.: +1-405-744-8867
| |
Collapse
|
20
|
Machado MG, Sencio V, Trottein F. Short-Chain Fatty Acids as a Potential Treatment for Infections: a Closer Look at the Lungs. Infect Immun 2021; 89:e0018821. [PMID: 34097474 PMCID: PMC8370681 DOI: 10.1128/iai.00188-21] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are the main metabolites produced by the gut microbiota via the fermentation of complex carbohydrates and fibers. Evidence suggests that SCFAs play a role in the control of infections through direct action both on microorganisms and on host signaling. This review summarizes the main microbicidal effects of SCFAs and discusses studies highlighting the effect of SCFAs in the virulence and viability of microorganisms. We also describe the diverse and complex modes of action of the SCFAs on the immune system in the face of infections with a specific focus on bacterial and viral respiratory infections. A growing body of evidence suggests that SCFAs protect against lung infections. Finally, we present potential strategies that may be leveraged to exploit the biological properties of SCFAs for increasing effectiveness and optimizing patient benefits.
Collapse
Affiliation(s)
- Marina Gomes Machado
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, ICB, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Valentin Sencio
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - François Trottein
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
21
|
Abhimanyu, Ontiveros CO, Guerra-Resendez RS, Nishiguchi T, Ladki M, Hilton IB, Schlesinger LS, DiNardo AR. Reversing Post-Infectious Epigenetic-Mediated Immune Suppression. Front Immunol 2021; 12:688132. [PMID: 34163486 PMCID: PMC8215363 DOI: 10.3389/fimmu.2021.688132] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/17/2021] [Indexed: 12/20/2022] Open
Abstract
The immune response must balance the pro-inflammatory, cell-mediated cytotoxicity with the anti-inflammatory and wound repair response. Epigenetic mechanisms mediate this balance and limit host immunity from inducing exuberant collateral damage to host tissue after severe and chronic infections. However, following treatment for these infections, including sepsis, pneumonia, hepatitis B, hepatitis C, HIV, tuberculosis (TB) or schistosomiasis, detrimental epigenetic scars persist, and result in long-lasting immune suppression. This is hypothesized to be one of the contributing mechanisms explaining why survivors of infection have increased all-cause mortality and increased rates of unrelated secondary infections. The mechanisms that induce epigenetic-mediated immune suppression have been demonstrated in-vitro and in animal models. Modulation of the AMP-activated protein kinase (AMPK)-mammalian target of rapamycin (mTOR), nuclear factor of activated T cells (NFAT) or nuclear receptor (NR4A) pathways is able to block or reverse the development of detrimental epigenetic scars. Similarly, drugs that directly modify epigenetic enzymes, such as those that inhibit histone deacetylases (HDAC) inhibitors, DNA hypomethylating agents or modifiers of the Nucleosome Remodeling and DNA methylation (NuRD) complex or Polycomb Repressive Complex (PRC) have demonstrated capacity to restore host immunity in the setting of cancer-, LCMV- or murine sepsis-induced epigenetic-mediated immune suppression. A third clinically feasible strategy for reversing detrimental epigenetic scars includes bioengineering approaches to either directly reverse the detrimental epigenetic marks or to modify the epigenetic enzymes or transcription factors that induce detrimental epigenetic scars. Each of these approaches, alone or in combination, have ablated or reversed detrimental epigenetic marks in in-vitro or in animal models; translational studies are now required to evaluate clinical applicability.
Collapse
Affiliation(s)
- Abhimanyu
- The Global Tuberculosis Program, William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Immigrant and Global Health, Baylor College of Medicine, Houston, TX, United States
| | - Carlos O Ontiveros
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, United States.,UT Health San Antonio, San Antonio, TX, United States
| | - Rosa S Guerra-Resendez
- Systems, Synthetic, and Physical Biology Graduate Program, Rice University, Houston, TX, United States
| | - Tomoki Nishiguchi
- The Global Tuberculosis Program, William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Immigrant and Global Health, Baylor College of Medicine, Houston, TX, United States
| | - Malik Ladki
- The Global Tuberculosis Program, William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Immigrant and Global Health, Baylor College of Medicine, Houston, TX, United States
| | - Isaac B Hilton
- Systems, Synthetic, and Physical Biology Graduate Program, Rice University, Houston, TX, United States.,Department of Bioengineering, Rice University, Houston, TX, United States.,Department of BioSciences, Rice University, Houston, TX, United States
| | - Larry S Schlesinger
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Andrew R DiNardo
- The Global Tuberculosis Program, William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Immigrant and Global Health, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
22
|
Bai J, Li Y, Li M, Tan S, Wu D. IL-37 As a Potential Biotherapeutics of Inflammatory Diseases. Curr Drug Targets 2021; 21:855-863. [PMID: 32348214 DOI: 10.2174/1389450121666200429114926] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/26/2022]
Abstract
Interleukin-37 (IL-37) was discovered as a new member of pro-inflammatory IL-1 superfamily. However, further studies suggested that IL-37 plays a critical anti-inflammatory role in innate and adaptive immunity. IL-37 may suppress the inflammatory process via intracellular SMAD family member 3 (SMAD3) and extracellular IL-18 Receptor alpha (IL-18Rα) signaling pathway, respectively. Meanwhile, the abnormal expression of IL-37 was observed in immune-mediated inflammatory diseases, such as inflammatory bowel disease, rheumatoid arthritis, atherosclerosis, systemic lupus erythematosus, asthma, and multiple sclerosis, which suggest IL-37 is a potential therapeutic target for these diseases. In this review, we summarize the anti-inflammatory mechanism of IL-37 and discuss the critical roles of IL-37 in the pathogenesis of these diseases. Further studies are required to confirm the effectiveness of IL-37 as a novel target for these inflammatory diseases.
Collapse
Affiliation(s)
- Junhui Bai
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China
| | - Yukun Li
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China
| | - Meixiang Li
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China
| | - Sijie Tan
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China
| | - Daichao Wu
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China
| |
Collapse
|
23
|
Sencio V, Barthelemy A, Tavares LP, Machado MG, Soulard D, Cuinat C, Queiroz-Junior CM, Noordine ML, Salomé-Desnoulez S, Deryuter L, Foligné B, Wahl C, Frisch B, Vieira AT, Paget C, Milligan G, Ulven T, Wolowczuk I, Faveeuw C, Le Goffic R, Thomas M, Ferreira S, Teixeira MM, Trottein F. Gut Dysbiosis during Influenza Contributes to Pulmonary Pneumococcal Superinfection through Altered Short-Chain Fatty Acid Production. Cell Rep 2021; 30:2934-2947.e6. [PMID: 32130898 DOI: 10.1016/j.celrep.2020.02.013] [Citation(s) in RCA: 232] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/13/2019] [Accepted: 02/04/2020] [Indexed: 02/07/2023] Open
Abstract
Secondary bacterial infections often complicate viral respiratory infections. We hypothesize that perturbation of the gut microbiota during influenza A virus (IAV) infection might favor respiratory bacterial superinfection. Sublethal infection with influenza transiently alters the composition and fermentative activity of the gut microbiota in mice. These changes are attributed in part to reduced food consumption. Fecal transfer experiments demonstrate that the IAV-conditioned microbiota compromises lung defenses against pneumococcal infection. In mechanistic terms, reduced production of the predominant short-chain fatty acid (SCFA) acetate affects the bactericidal activity of alveolar macrophages. Following treatment with acetate, mice colonized with the IAV-conditioned microbiota display reduced bacterial loads. In the context of influenza infection, acetate supplementation reduces, in a free fatty acid receptor 2 (FFAR2)-dependent manner, local and systemic bacterial loads. This translates into reduced lung pathology and improved survival rates of double-infected mice. Lastly, pharmacological activation of the SCFA receptor FFAR2 during influenza reduces bacterial superinfection.
Collapse
Affiliation(s)
- Valentin Sencio
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Adeline Barthelemy
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Luciana P Tavares
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marina G Machado
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France; Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daphnée Soulard
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Céline Cuinat
- Micalis Institute, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | | | - Marie-Louise Noordine
- Micalis Institute, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Sophie Salomé-Desnoulez
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Lucie Deryuter
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Benoit Foligné
- Université de Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), CHU Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France
| | | | - Benoit Frisch
- Centre National de la Recherche Scientifique, Université de Strasbourg, Faculté de Pharmacie, 67400 Illkirch, France
| | - Angelica T Vieira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Christophe Paget
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, G12 8QQ Glasgow, Scotland, UK
| | - Trond Ulven
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Isabelle Wolowczuk
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Christelle Faveeuw
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Ronan Le Goffic
- Molecular Virology and Immunology, Institut National de la Recherche Agronomique, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Muriel Thomas
- Micalis Institute, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | | | - Mauro M Teixeira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - François Trottein
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France.
| |
Collapse
|
24
|
DiNardo AR, Nishiguchi T, Grimm SL, Schlesinger LS, Graviss EA, Cirillo JD, Coarfa C, Mandalakas AM, Heyckendorf J, Kaufmann SHE, Lange C, Netea MG, Van Crevel R. Tuberculosis endotypes to guide stratified host-directed therapy. MED 2021; 2:217-232. [PMID: 34693385 DOI: 10.1016/j.medj.2020.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
There is hope that host-directed therapy (HDT) for Tuberculosis (TB) can either shorten treatment duration, help cure drug resistant disease or limit the immunopathology. Many candidate HDT drugs have been proposed, however solid evidence only exists for a few select patient groups. The clinical presentation of TB is variable, with differences in severity, tissue pathology, and bacillary burden. TB clinical phenotypes likely determine the potential benefit of HDT. Underlying TB clinical phenotypes, there are TB "endotypes," defined as distinct molecular profiles, with specific metabolic, epigenetic, transcriptional, and immune phenotypes. TB endotypes can be characterized by either immunodeficiency or pathologic excessive inflammation. Additional factors, like comorbidities (HIV, diabetes, helminth infection), structural lung disease or Mycobacterial virulence also drive TB endotypes. Precise disease phenotyping, combined with in-depth immunologic and molecular profiling and multimodal omics integration, can identify TB endotypes, guide endotype-specific HDT, and improve TB outcomes, similar to advances in cancer medicine.
Collapse
Affiliation(s)
- Andrew R DiNardo
- The Global Tuberculosis Program, Texas Children's Hospital, Immigrant and Global Health, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Tomoki Nishiguchi
- The Global Tuberculosis Program, Texas Children's Hospital, Immigrant and Global Health, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Sandra L Grimm
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Molecular and Cellular Biology, Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | | | - Edward A Graviss
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Jeffrey D Cirillo
- Department of Microbial and Molecular Pathogenesis, Texas A&M College of Medicine, Bryan, TX, USA
| | - Cristian Coarfa
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Molecular and Cellular Biology, Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Anna M Mandalakas
- The Global Tuberculosis Program, Texas Children's Hospital, Immigrant and Global Health, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Jan Heyckendorf
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany.,German Center for Infection Research (DZIF) Clinical Tuberculosis Unit, Borstel, Germany.,Respiratory Medicine & International Health, University of Lübeck, Lü beck, Germany
| | - Stefan H E Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany.,Hagler Institute for Advanced Study, Texas A&M University, College Station, TX, USA.,Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077 Gö ttingen, Germany
| | - Christoph Lange
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany.,German Center for Infection Research (DZIF) Clinical Tuberculosis Unit, Borstel, Germany.,Respiratory Medicine & International Health, University of Lübeck, Lü beck, Germany
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Immunology and Metabolism, Life & Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Reinout Van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
25
|
Pimentel PMDO, de Assis DRR, Gualdrón-Lopez M, Barroso A, Brant F, Leite PG, de Lima Oliveira BC, Esper L, McKinnie SMK, Vederas JC, do Nascimento Cordeiro M, Dos Reis PVM, Teixeira MM, de Castro Pimenta AM, Borges MH, de Lima ME, Machado FS. Tityus serrulatus scorpion venom as a potential drug source for Chagas' disease: Trypanocidal and immunomodulatory activity. Clin Immunol 2021; 226:108713. [PMID: 33711450 DOI: 10.1016/j.clim.2021.108713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/19/2021] [Accepted: 03/06/2021] [Indexed: 11/19/2022]
Abstract
Current chemical therapies for Chagas Disease (CD) lack ability to clear Trypanosoma cruzi (Tc) parasites and cause severe side effects, making search for new strategies extremely necessary. We evaluated the action of Tityus serrulatus venom (TsV) components during Tc infection. TsV treatment increased nitric oxide and pro-inflammatory cytokine production by Tc-infected macrophages (MØ), decreased intracellular parasite replication and trypomastigotes release, also triggering ERK1/2, JNK1/2 and p38 activation. Ts7 demonstrated the highest anti-Tc activity, inducing high levels of TNF and IL-6 in infected MØ. TsV/Ts7 presented synergistic effect on p38 activation when incubated with Tc antigen. KPP-treatment of MØ also decreased trypomastigotes releasing, partially due to p38 activation. TsV/Ts7-pre-incubation of Tc demonstrated a direct effect on parasite decreasing MØ-trypomastigotes releasing. In vivo KPP-treatment of Tc-infected mice resulted in decreased parasitemia. Summarizing, this study opens perspectives for new bioactive molecules as CD-therapeutic treatment, demonstrating the TsV/Ts7/KPP-trypanocidal and immunomodulatory activity during Tc infection.
Collapse
Affiliation(s)
| | - Diego Rodney Rodrigues de Assis
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Melisa Gualdrón-Lopez
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Andréia Barroso
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fátima Brant
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paulo Gaio Leite
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bruno Cabral de Lima Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lisia Esper
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Shaun M K McKinnie
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada; Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA, USA
| | - John C Vederas
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Pablo Victor Mendes Dos Reis
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adriano Monteiro de Castro Pimenta
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Maria Elena de Lima
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Santa Casa BH: Instituto de Ensino e Pesquisa, Belo Horizonte, Brazil
| | - Fabiana Simão Machado
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
26
|
Ramos-Espinosa O, Mata-Espinosa D, Francisco-Cruz A, López-Torres MO, Hernández-Bazán S, Barrios-Payán J, Marquina-Castillo B, Carretero M, Del Río M, Hernández-Pando R. Immunotherapeutic effect of adenovirus encoding antimicrobial peptides in experimental pulmonary tuberculosis. J Leukoc Biol 2021; 110:951-963. [PMID: 33682193 DOI: 10.1002/jlb.4ma0920-627r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/14/2020] [Accepted: 01/02/2021] [Indexed: 11/11/2022] Open
Abstract
As components of the innate immune response, antimicrobial peptides (AMPs) efficiently contribute to infection control and maintenance of a latent state in pulmonary tuberculosis (TB). As a therapeutic strategy, the administration of recombinant AMPs could be limited by enzymatic degradation and high production costs. Likewise, strategies based on the induction of AMPs have generated controversial results. In this study, 2 recombinant type-5 adenoviruses (Ad) expressing the human β-defensin 3 (HβD3) or cathelicidin (LL37) were assessed in a murine pulmonary TB model. Mice infected with either a high dose of a drug-sensitive (H37Rv) or a multidrug-resistant (MDR) strain of Mycobacterium tuberculosis (Mtb) were treated with a single administration of AdHβD3, AdLL37, AdGFP (control vector expressing a green fluorescent protein), or saline solution (SS). Lungs were obtained to determine the bacterial burden, histologic damage, and cytokine expression at different time points. Mice treated with AdHβD3 or AdLL37 showed significantly lower bacterial load and pneumonia, and higher proinflammatory cytokine expression than the control groups AdGFP and SS. A synergistic therapeutic effect could be observed when first- or second-line antibiotics (ABs) were administered with adenoviral therapy in animals infected with H37Rv or MDR strains, respectively. Adenovirus-delivered AMP's administration constitutes a promising adjuvant therapy for current anti-TB drugs by enhancing a protective immune response and potentially reducing current AB regimes' duration.
Collapse
Affiliation(s)
- Octavio Ramos-Espinosa
- Experimental Pathology Section, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Dulce Mata-Espinosa
- Experimental Pathology Section, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Alejandro Francisco-Cruz
- Department of Translational Molecular Pathology, Division of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Manuel Othoniel López-Torres
- Experimental Pathology Section, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sujhey Hernández-Bazán
- Experimental Pathology Section, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jorge Barrios-Payán
- Experimental Pathology Section, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Brenda Marquina-Castillo
- Experimental Pathology Section, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Marta Carretero
- Epithelial Damage, Repair and Tissue Engineering, Ciemat-Fundación Marcelino Botín, Complutense University, Madrid, Spain.,IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Marcela Del Río
- Epithelial Damage, Repair and Tissue Engineering, Ciemat-Fundación Marcelino Botín, Complutense University, Madrid, Spain.,IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Rogelio Hernández-Pando
- Experimental Pathology Section, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
27
|
Chauhan A, Kumar M, Kumar A, Kanchan K. Comprehensive review on mechanism of action, resistance and evolution of antimycobacterial drugs. Life Sci 2021; 274:119301. [PMID: 33675895 DOI: 10.1016/j.lfs.2021.119301] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/14/2021] [Accepted: 02/24/2021] [Indexed: 01/04/2023]
Abstract
Tuberculosis is one of the deadliest infectious diseases existing in the world since ancient times and still possesses serious threat across the globe. Each year the number of cases increases due to high drug resistance shown by Mycobacterium tuberculosis (Mtb). Available antimycobacterial drugs have been classified as First line, Second line and Third line antibiotics depending on the time of their discoveries and their effectiveness in the treatment. These antibiotics have a broad range of targets ranging from cell wall to metabolic processes and their non-judicious and uncontrolled usage in the treatment for years has created a significant problem called multi-drug resistant (MDR) tuberculosis. In this review, we have summarized the mechanism of action of all the classified antibiotics currently in use along with the resistance mechanisms acquired by Mtb. We have focused on the new drug candidates/repurposed drugs, and drug in combinations, which are in clinical trials for either treating the MDR tuberculosis more effectively or involved in reducing the time required for the chemotherapy of drug sensitive TB. This information is not discussed very adequately on a single platform. Additionally, we have discussed the recent technologies that are being used to discover novel resistance mechanisms acquired by Mtb and for exploring novel drugs. The story of intrinsic resistance mechanisms and evolution in Mtb is far from complete. Therefore, we have also discussed intrinsic resistance mechanisms of Mtb and their evolution with time, emphasizing the hope for the development of novel antimycobacterial drugs for effective therapy of tuberculosis.
Collapse
Affiliation(s)
- Aditi Chauhan
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida 201313, India
| | - Manoj Kumar
- Amity Food and Agriculture Foundation, Amity University Uttar Pradesh, Noida 201313, India
| | - Awanish Kumar
- Department of Bio Technology, National Institute of Technology, Raipur, India
| | - Kajal Kanchan
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida 201313, India.
| |
Collapse
|
28
|
Campo M, Heater S, Peterson GJ, Simmons JD, Skerrett SJ, Mayanja-Kizza H, Stein CM, Boom WH, Hawn TR. HDAC3 inhibitor RGFP966 controls bacterial growth and modulates macrophage signaling during Mycobacterium tuberculosis infection. Tuberculosis (Edinb) 2021; 127:102062. [PMID: 33639591 PMCID: PMC8650124 DOI: 10.1016/j.tube.2021.102062] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 01/28/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022]
Abstract
RATIONALE Host-directed therapeutics for Mycobacterium tuberculosis (Mtb) offer potential strategies for combatting antibiotic resistance and for killing non-replicating bacilli. Phenylbutyrate, a partially selective histone-deacetylase (HDAC) inhibitor, was previously shown to control Mtb growth and alter macrophage inflammatory pathways at 2-4 mM concentrations. OBJECTIVE To identify a more potent and selective HDAC inhibitor that modulates macrophage responses to mycobacteria and has direct antibacterial effects against Mtb. METHODS We used cellular approaches to characterize the role of pharmacologic inhibition of HDAC3 on Mtb growth and Mtb-induced peripheral and alveolar macrophage immune functions. MEASUREMENTS AND MAIN RESULTS RGFP966, an HDAC3 inhibitor, controlled Mtb, BCG and M. avium growth directly in broth culture and in human peripheral blood monocyte-derived and alveolar macrophages with an MIC50 of approximately 5-10 μM. In contrast, RGFP966 did not inhibit growth of several other intracellular and extracellular bacteria. We also found that RGFP966 modulated macrophage pro-inflammatory cytokine secretion in response to Mtb infection with decreased IL6 and TNF secretion. CONCLUSIONS We identified a potent and selective small molecule inhibitor of HDAC3 with direct antimicrobial activity against Mtb and modulation of macrophage signaling pathways.
Collapse
MESH Headings
- Acrylamides/pharmacology
- Adolescent
- Adult
- Antitubercular Agents/pharmacology
- Cells, Cultured
- Cytokines/metabolism
- Female
- Histone Deacetylase Inhibitors/pharmacology
- Histone Deacetylases/metabolism
- Host-Pathogen Interactions
- Humans
- Immunity, Innate/drug effects
- Inflammation Mediators/metabolism
- Macrophages, Alveolar/drug effects
- Macrophages, Alveolar/enzymology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/microbiology
- Male
- Middle Aged
- Mycobacterium tuberculosis/drug effects
- Mycobacterium tuberculosis/growth & development
- Mycobacterium tuberculosis/immunology
- Phenylenediamines/pharmacology
- Signal Transduction
- Tuberculosis, Pulmonary/drug therapy
- Tuberculosis, Pulmonary/enzymology
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/microbiology
- Young Adult
Collapse
Affiliation(s)
- Monica Campo
- Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Sarah Heater
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | | | - Jason D Simmons
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Shawn J Skerrett
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Harriet Mayanja-Kizza
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Catherine M Stein
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Medicine, School of Medicine, Makerere University and Mulago Hospital, Kampala, Uganda
| | - W Henry Boom
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Thomas R Hawn
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
29
|
Rao Muvva J, Ahmed S, Rekha RS, Kalsum S, Groenheit R, Schön T, Agerberth B, Bergman P, Brighenti S. Immunomodulatory Agents Combat Multidrug-Resistant Tuberculosis by Improving Antimicrobial Immunity. J Infect Dis 2021; 224:332-344. [PMID: 33606878 PMCID: PMC8280489 DOI: 10.1093/infdis/jiab100] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 02/13/2021] [Indexed: 12/15/2022] Open
Abstract
Background Multidrug-resistant (MDR) tuberculosis has low treatment success rates, and new treatment strategies are needed. We explored whether treatment with active vitamin D3 (vitD) and phenylbutyrate (PBA) could improve conventional chemotherapy by enhancing immune-mediated eradication of Mycobacterium tuberculosis. Methods A clinically relevant model was used consisting of human macrophages infected with M. tuberculosis isolates (n = 15) with different antibiotic resistance profiles. The antimicrobial effect of vitD+PBA, was tested together with rifampicin or isoniazid. Methods included colony-forming units (intracellular bacterial growth), messenger RNA expression analyses (LL-37, β-defensin, nitric oxide synthase, and dual oxidase 2), RNA interference (LL-37-silencing in primary macrophages), and Western blot analysis and confocal microscopy (LL-37 and LC3 protein expression). Results VitD+PBA inhibited growth of clinical MDR tuberculosis strains in human macrophages and strengthened intracellular growth inhibition of rifampicin and isoniazid via induction of the antimicrobial peptide LL-37 and LC3-dependent autophagy. Gene silencing of LL-37 expression enhanced MDR tuberculosis growth in vitD+PBA–treated macrophages. The combination of vitD+PBA and isoniazid were as effective in reducing intracellular MDR tuberculosis growth as a >125-fold higher dose of isoniazid alone, suggesting potent additive effects of vitD+PBA with isoniazid. Conclusions Immunomodulatory agents that trigger multiple immune pathways can strengthen standard MDR tuberculosis treatment and contribute to next-generation individualized treatment options for patients with difficult-to-treat pulmonary tuberculosis.
Collapse
Affiliation(s)
- Jagadeeswara Rao Muvva
- Center for Infectious Medicine (CIM), Department of Medicine, ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Sultan Ahmed
- Clinical Microbiology, Department of Laboratory Medicine (Labmed), ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Rokeya Sultana Rekha
- Clinical Microbiology, Department of Laboratory Medicine (Labmed), ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Sadaf Kalsum
- Center for Infectious Medicine (CIM), Department of Medicine, ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Ramona Groenheit
- Department of Microbiology, Public Health Agency of Sweden , Solna, Sweden
| | - Thomas Schön
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, and Department of Clinical Microbiology and Infectious Diseases, Kalmar County Hospital, Kalmar, Sweden
| | - Birgitta Agerberth
- Clinical Microbiology, Department of Laboratory Medicine (Labmed), ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Peter Bergman
- Clinical Microbiology, Department of Laboratory Medicine (Labmed), ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Susanna Brighenti
- Center for Infectious Medicine (CIM), Department of Medicine, ANA Futura, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
30
|
Kilinç G, Saris A, Ottenhoff THM, Haks MC. Host-directed therapy to combat mycobacterial infections. Immunol Rev 2021; 301:62-83. [PMID: 33565103 PMCID: PMC8248113 DOI: 10.1111/imr.12951] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 12/27/2020] [Indexed: 12/27/2022]
Abstract
Upon infection, mycobacteria, such as Mycobacterium tuberculosis (Mtb) and nontuberculous mycobacteria (NTM), are recognized by host innate immune cells, triggering a series of intracellular processes that promote mycobacterial killing. Mycobacteria, however, have developed multiple counter‐strategies to persist and survive inside host cells. By manipulating host effector mechanisms, including phagosome maturation, vacuolar escape, autophagy, antigen presentation, and metabolic pathways, pathogenic mycobacteria are able to establish long‐lasting infection. Counteracting these mycobacteria‐induced host modifying mechanisms can be accomplished by host‐directed therapeutic (HDT) strategies. HDTs offer several major advantages compared to conventional antibiotics: (a) HDTs can be effective against both drug‐resistant and drug‐susceptible bacteria, as well as potentially dormant mycobacteria; (b) HDTs are less likely to induce bacterial drug resistance; and (c) HDTs could synergize with, or shorten antibiotic treatment by targeting different pathways. In this review, we will explore host‐pathogen interactions that have been identified for Mtb for which potential HDTs impacting both innate and adaptive immunity are available, and outline those worthy of future research. We will also discuss possibilities to target NTM infection by HDT, although current knowledge regarding host‐pathogen interactions for NTM is limited compared to Mtb. Finally, we speculate that combinatorial HDT strategies can potentially synergize to achieve optimal mycobacterial host immune control.
Collapse
Affiliation(s)
- Gül Kilinç
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Anno Saris
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
31
|
Chai Q, Wang L, Liu CH, Ge B. New insights into the evasion of host innate immunity by Mycobacterium tuberculosis. Cell Mol Immunol 2020; 17:901-913. [PMID: 32728204 PMCID: PMC7608469 DOI: 10.1038/s41423-020-0502-z] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/22/2020] [Indexed: 12/26/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is an extremely successful intracellular pathogen that causes tuberculosis (TB), which remains the leading infectious cause of human death. The early interactions between Mtb and the host innate immune system largely determine the establishment of TB infection and disease development. Upon infection, host cells detect Mtb through a set of innate immune receptors and launch a range of cellular innate immune events. However, these innate defense mechanisms are extensively modulated by Mtb to avoid host immune clearance. In this review, we describe the emerging role of cytosolic nucleic acid-sensing pathways at the host-Mtb interface and summarize recently revealed mechanisms by which Mtb circumvents host cellular innate immune strategies such as membrane trafficking and integrity, cell death and autophagy. In addition, we discuss the newly elucidated strategies by which Mtb manipulates the host molecular regulatory machinery of innate immunity, including the intranuclear regulatory machinery, the ubiquitin system, and cellular intrinsic immune components. A better understanding of innate immune evasion mechanisms adopted by Mtb will provide new insights into TB pathogenesis and contribute to the development of more effective TB vaccines and therapies.
Collapse
Affiliation(s)
- Qiyao Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, 100101, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Lin Wang
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 200433, Shanghai, China
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, 100101, Beijing, China. .,Savaid Medical School, University of Chinese Academy of Sciences, 101408, Beijing, China.
| | - Baoxue Ge
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 200433, Shanghai, China.
| |
Collapse
|
32
|
Bergman P, Raqib R, Rekha RS, Agerberth B, Gudmundsson GH. Host Directed Therapy Against Infection by Boosting Innate Immunity. Front Immunol 2020; 11:1209. [PMID: 32595649 PMCID: PMC7304486 DOI: 10.3389/fimmu.2020.01209] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/15/2020] [Indexed: 12/18/2022] Open
Abstract
The innate immune system constitutes the first line of defense against invading pathogens, regulating the normal microbiota and contributes to homeostasis. Today we have obtained detailed knowledge on receptors, signaling pathways, and effector molecules of innate immunity. Our research constellation has focused on ways to induce the expression of antimicrobial peptides (AMPs), the production of oxygen species (ROS and NO), and to activate autophagy, during the last two decades. These innate effectors, with different mechanisms of action, constitute a powerful defense armament in phagocytes and in epithelial cells. Innate immunity does not only protect the host from invading pathogens, but also regulates the composition of the microbiota, which is an area of intense research. Notably, some virulent bacteria have the capacity to downregulate innate defenses and can thereby cause invasive disease. Understanding the detailed mechanisms behind pathogen-mediated suppression of innate effectors are currently in progress. This information can be of importance for the development of novel treatments based on counteraction of the downregulation; we have designated this type of treatment as host directed therapy (HDT). The concept to boost innate immunity may be particularly relevant as many pathogens are developing resistance against classical antibiotics. Many pathogens that are resistant to antibiotics are sensitive to the endogenous effectors included in early host defenses, which contain multiple effectors working in cooperation to control infections. Here, we review recent data related to downregulation of AMPs by pathogenic bacteria, induction of innate effector mechanisms, including cytokine-mediated effects, repurposed drugs and the role of antibiotics as direct modulators of host responses. These findings can form a platform for the development of novel treatment strategies against infection and/or inflammation.
Collapse
Affiliation(s)
- Peter Bergman
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,The Immunodeficiency Unit, Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Rubhana Raqib
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Rokeya Sultana Rekha
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Birgitta Agerberth
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gudmundur H Gudmundsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Biomedical Center, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
33
|
Chai Q, Lu Z, Liu CH. Host defense mechanisms against Mycobacterium tuberculosis. Cell Mol Life Sci 2020; 77:1859-1878. [PMID: 31720742 PMCID: PMC11104961 DOI: 10.1007/s00018-019-03353-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/30/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022]
Abstract
Tuberculosis (TB), which is caused by Mycobacterium tuberculosis (Mtb), remains the leading cause of death worldwide from a single infectious pathogen. Mtb is a paradigmatic intracellular pathogen that primarily invades the lungs after host inhalation of bacteria-containing droplets via the airway. However, the majority of Mtb-exposed individuals can spontaneously control the infection by virtue of a robust immune defense system. The mucosal barriers of the respiratory tract shape the first-line defense against Mtb through various mucosal immune responses. After arriving at the alveoli, the surviving mycobacteria further encounter a set of host innate immune cells that exert multiple cellular bactericidal functions. Adaptive immunity, predominantly mediated by a range of different T cell and B cell subsets, is subsequently activated and participates in host anti-mycobacterial defense. During Mtb infection, host bactericidal immune responses are exquisitely adjusted and balanced by multifaceted mechanisms, including genetic and epigenetic regulation, metabolic regulation and neuroendocrine regulation, which are indispensable for maintaining host immune efficiency and avoiding excessive tissue injury. A better understanding of the integrated and equilibrated host immune defense system against Mtb will contribute to the development of rational TB treatment regimens especially novel host-directed therapeutics.
Collapse
Affiliation(s)
- Qiyao Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Zhe Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
34
|
Young C, Walzl G, Du Plessis N. Therapeutic host-directed strategies to improve outcome in tuberculosis. Mucosal Immunol 2020; 13:190-204. [PMID: 31772320 PMCID: PMC7039813 DOI: 10.1038/s41385-019-0226-5] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/15/2019] [Accepted: 10/23/2019] [Indexed: 02/04/2023]
Abstract
Bacille Calmette-Guérin (BCG) is the only licenced tuberculosis (TB) vaccine, but has limited efficacy against pulmonary TB disease development and modest protection against extrapulmonary TB. Preventative antibiotic treatment for Mycobacterium tuberculosis (Mtb) infections in high-prevalence settings is unfeasible due to unclear treatment durability, drug toxicity, logistical constraints related to directly observed treatment strategy (DOTS) and the lengthy treatment protocols. Together, these factors promote non-adherence, contributing to relapse and establishment of drug-resistant Mtb strains. Although antibiotic treatment of drug-susceptible Mtb is generally effective, drug-resistant TB has a treatment efficacy below 50% and can, in a proportion, develop into progressive, untreatable disease. Other immune compromising co-infections and/or co-morbidities require more complex prevention/treatment approaches, posing huge financial burdens to national health services. Novel TB treatment strategies, such as host-directed therapeutics, are required to complement pathogen-targeted approaches. Pre-clinical studies have highlighted promising candidates that enhance endogenous pathways and/or limit destructive host responses. This review discusses promising pre-clinical candidates and forerunning compounds at advanced stages of clinical investigation in TB host-directed therapeutic (HDT) efficacy trials. Such approaches are rationalized to improve outcome in TB and shorten treatment strategies.
Collapse
Affiliation(s)
- C Young
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology/DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - G Walzl
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology/DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - N Du Plessis
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology/DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
35
|
Moreira JD, Koch BEV, van Veen S, Walburg KV, Vrieling F, Mara Pinto Dabés Guimarães T, Meijer AH, Spaink HP, Ottenhoff THM, Haks MC, Heemskerk MT. Functional Inhibition of Host Histone Deacetylases (HDACs) Enhances in vitro and in vivo Anti-mycobacterial Activity in Human Macrophages and in Zebrafish. Front Immunol 2020; 11:36. [PMID: 32117228 PMCID: PMC7008710 DOI: 10.3389/fimmu.2020.00036] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/08/2020] [Indexed: 12/27/2022] Open
Abstract
The rapid and persistent increase of drug-resistant Mycobacterium tuberculosis (Mtb) infections poses increasing global problems in combatting tuberculosis (TB), prompting for the development of alternative strategies including host-directed therapy (HDT). Since Mtb is an intracellular pathogen with a remarkable ability to manipulate host intracellular signaling pathways to escape from host defense, pharmacological reprogramming of the immune system represents a novel, potentially powerful therapeutic strategy that should be effective also against drug-resistant Mtb. Here, we found that host-pathogen interactions in Mtb-infected primary human macrophages affected host epigenetic features by modifying histone deacetylase (HDAC) transcriptomic levels. In addition, broad spectrum inhibition of HDACs enhanced the antimicrobial response of both pro-inflammatory macrophages (Mϕ1) and anti-inflammatory macrophages (Mϕ2), while selective inhibition of class IIa HDACs mainly decreased bacterial outgrowth in Mϕ2. Moreover, chemical inhibition of HDAC activity during differentiation polarized macrophages into a more bactericidal phenotype with a concomitant decrease in the secretion levels of inflammatory cytokines. Importantly, in vivo chemical inhibition of HDAC activity in Mycobacterium marinum-infected zebrafish embryos, a well-characterized animal model for tuberculosis, significantly reduced mycobacterial burden, validating our in vitro findings in primary human macrophages. Collectively, these data identify HDACs as druggable host targets for HDT against intracellular Mtb.
Collapse
Affiliation(s)
- Jôsimar D Moreira
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.,Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Bjørn E V Koch
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Suzanne van Veen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Kimberley V Walburg
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Frank Vrieling
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tânia Mara Pinto Dabés Guimarães
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Herman P Spaink
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Matthias T Heemskerk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
36
|
Host-Directed Therapy as a Novel Treatment Strategy to Overcome Tuberculosis: Targeting Immune Modulation. Antibiotics (Basel) 2020; 9:antibiotics9010021. [PMID: 31936156 PMCID: PMC7168302 DOI: 10.3390/antibiotics9010021] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/25/2019] [Accepted: 01/04/2020] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB) is one of the leading causes of mortality and morbidity, particularly in developing countries, presenting a major threat to the public health. The currently recommended long term treatment regimen with multiple antibiotics is associated with poor patient compliance, which in turn, may contribute to the emergence of multi-drug resistant TB (MDR-TB). The low global treatment efficacy of MDR-TB has highlighted the necessity to develop novel treatment options. Host-directed therapy (HDT) together with current standard anti-TB treatments, has gained considerable interest, as HDT targets novel host immune mechanisms. These immune mechanisms would otherwise bypass the antibiotic bactericidal targets to kill Mycobacterium tuberculosis (Mtb), which may be mutated to cause antibiotic resistance. Additionally, host-directed therapies against TB have been shown to be associated with reduced lung pathology and improved disease outcome, most likely via the modulation of host immune responses. This review will provide an update of host-directed therapies and their mechanism(s) of action against Mycobacterium tuberculosis.
Collapse
|
37
|
Host-Targeted Therapeutics against Multidrug Resistant Intracellular Staphylococcus aureus. Antibiotics (Basel) 2019; 8:antibiotics8040241. [PMID: 31795127 PMCID: PMC6963206 DOI: 10.3390/antibiotics8040241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus is a facultative intracellular pathogen that invades and replicates within many types of human cells. S. aureus has shown to rapidly overcome traditional antibiotherapy by developing multidrug resistance. Furthermore, intracellular S. aureus is protected from the last-resort antibiotics—vancomycin, daptomycin, and linezolid—as they are unable to achieve plasma concentrations sufficient for intracellular killing. Therefore, there is an urgent need to develop novel anti-infective therapies against S. aureus infections. Here, we review the current state of the field and highlight the exploitation of host-directed approaches as a promising strategy going forward.
Collapse
|
38
|
Kaipilyawar V, Salgame P. Infection resisters: targets of new research for uncovering natural protective immunity against Mycobacterium tuberculosis. F1000Res 2019; 8. [PMID: 31602294 PMCID: PMC6774050 DOI: 10.12688/f1000research.19805.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2019] [Indexed: 12/17/2022] Open
Abstract
“Infection resisters” are broadly defined as individuals who despite significant exposure to
Mycobacterium tuberculosis remain persistently unreactive to conventional detection assays, suggesting that they remain uninfected or rapidly clear their infection early on following exposure. In this review, we highlight recent studies that point to underlying host immune mechanisms that could mediate this natural resistance. We also illustrate some additional avenues that are likely to be differently modulated in resisters and possess the potential to be targeted, ranging from early mycobacterial sensing leading up to subsequent killing. Emerging research in this area can be harnessed to provide valuable insights into the development of novel therapeutic and vaccine strategies against
M. tuberculosis.
Collapse
Affiliation(s)
- Vaishnavi Kaipilyawar
- Center for Emerging Pathogens, Rutgers-New Jersey Medical School, International Center for Public Health, 225 Warren St, Newark, NJ, 07103, USA
| | - Padmini Salgame
- Center for Emerging Pathogens, Rutgers-New Jersey Medical School, International Center for Public Health, 225 Warren St, Newark, NJ, 07103, USA
| |
Collapse
|
39
|
Montoya D, Mehta M, Ferguson BG, Teles RMB, Krutzik SR, Cruz D, Pellegrini M, Modlin RL. Plasticity of antimicrobial and phagocytic programs in human macrophages. Immunology 2019; 156:164-173. [PMID: 30357820 PMCID: PMC6328994 DOI: 10.1111/imm.13013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/01/2018] [Indexed: 01/03/2023] Open
Abstract
Macrophage (MΦ) polarization is triggered during the innate immune response to defend against microbial pathogens, but can also contribute to disease pathogenesis. In a previous study, we found that interleukin-15 (IL-15) -derived classically activated macrophages (M1 MΦ) have enhanced antimicrobial activity, whereas IL-10-derived alternatively activated macrophages (M2 MΦ) were highly phagocytic but lacked antimicrobial activity. Given that the ability to modulate MΦ polarization from M2 MΦ to M1 MΦ may promote a more effective immune response to infection, we investigated the plasticity of these MΦ programs. Addition of IL-10 to M1 MΦ induced M2-like MΦ, but IL-15 had little effect on M2 MΦ. We determined the set of immune receptors that are present on M2 MΦ, elucidating two candidates for inducing plasticity of M2 MΦ, Toll-like receptor 1 (TLR1) and interferonγ (IFN-γ) receptor 1. Stimulation of M2 MΦ with TLR2/1 ligand (TLR2/1L) or IFN-γ alone was not sufficient to alter M2 MΦ phenotype or function. However, co-addition of TLR2/1L and IFN-γ re-educated M2 MΦ towards the M1 MΦ phenotype, with a decrease in the phagocytosis of lipids and mycobacteria, as well as recovery of the vitamin-D-dependent antimicrobial pathway compared with M2 MΦ maintained in polarizing conditions. Similarly, treatment of M2 MΦ with both TLR2/1L and anti-IL-10 neutralizing antibodies led to polarization to the M1-like MΦ phenotype and function. Together, our data demonstrate an approach to induce MΦ plasticity that provides the potential for re-educating MΦ function in human mycobacterial disease to promote host defense and limit pathogenesis.
Collapse
Affiliation(s)
- Dennis Montoya
- Department of Molecular, Cell, and Developmental BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Manali Mehta
- Division of DermatologyDepartment of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | | | - Rosane M. B. Teles
- Division of DermatologyDepartment of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Stephan R. Krutzik
- Division of DermatologyDepartment of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Daniel Cruz
- Division of CardiologyDepartment of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Robert L. Modlin
- Division of DermatologyDepartment of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
40
|
Dou X, Han J, Ma Q, Cheng B, Shan A, Gao N, Yang Y. TLR2/4-mediated NF-κB pathway combined with the histone modification regulates β-defensins and interleukins expression by sodium phenyl butyrate in porcine intestinal epithelial cells. Food Nutr Res 2018; 62:1493. [PMID: 30574051 PMCID: PMC6294838 DOI: 10.29219/fnr.v62.1493] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/18/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022] Open
Abstract
Background Host defense peptides (HDPs) possess direct antibacterial, antineoplastic, and immunomodulatory abilities, playing a vital role in innate immunity. Dietary-regulated HDP holds immense potential as a novel pathway for preventing infection. Objective In this study, we examined the regulation mechanism of HDPs (pEP2C, pBD-1, and pBD-3) and cytokines (IL-8 and IL-18) expression by sodium phenylbutyrate (PBA). Design The effects of PBA on HDP induction and the mechanism involved were studied in porcine intestinal epithelial cell lines (IPEC J2). Results In this study, the results showed that HDPs (pEP2C, pBD-1, and pBD-3) and cytokines (IL-8 and IL-18) expression was increased significantly upon stimulation with PBA in IPEC J2 cells. Furthermore, toll-like receptor 2 (TLR2) and TLR4 were required for the PBA-mediated upregulation of the HDPs. This process occurred and further activated the NF-κB pathway via the phosphorylation of p65 and an IκB α synthesis delay. Meanwhile, histone deacetylase (HDAC) inhibition and an increased phosphorylation of histone H3 on serine S10 also occurred in PBA-induced HDP expression independently with TLR2 and TLR4. Furthermore, p38-MAPK suppressed PBA-induced pEP2C, pBD-1 pBD-3, IL-8, and IL-18 expression, but ERK1/2 failed to abolish the regulation of pBD-3, IL-8, and IL-18. Moreover, epidermal growth factor receptor (EGFR) is involved in PBA-mediated HDP regulation. Conclusions We concluded that PBA induced HDP and cytokine increases but did not cause an excessive pro-inflammatory response, which proceeded through the TLR2 and TLR4-NF-κB pathway and histone modification in IPEC J2 cells.
Collapse
Affiliation(s)
- Xiujing Dou
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Junlan Han
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Qiuyuan Ma
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Baojing Cheng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Nan Gao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Yu Yang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
41
|
Wang X, Zhang M, Jiang N, Zhang A. Sodium Phenylbutyrate Ameliorates Inflammatory Response Induced by Staphylococcus aureus Lipoteichoic Acid via Suppressing TLR2/NF-κB/NLRP3 Pathways in MAC-T Cells. Molecules 2018; 23:molecules23123056. [PMID: 30469547 PMCID: PMC6321250 DOI: 10.3390/molecules23123056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 01/01/2023] Open
Abstract
This study aimed to investigate the anti-inflammatory properties of sodium phenylbutyrate (SPB) against Staphylococcus aureus (S. aureus) lipoteichoic acid (LTA)-stimulated bovine mammary alveolar (MAC-T) cells. Quantitative PCR was performed to examine the effect of SPB on inflammatory cytokines and host defense peptide (HDP) gene expression. Western blot wanalysis was used to detect the effect of SPB on the TLR2/NF-κB/NLRP3 signaling pathway. The results showed that SPB significantly suppressed the expression of TNF-α, IL-1β, IL-6; meanwhile, the markedly decreased expression of LTA-stimulated TLR2, NLRP3, ASC, caspase-1, and IL-1β, and the inhibited IkBα and p65 phosphorylation were also observed. However, increased TAP and Bac5 expression in LTA-stimulated MAC-T cells was further detected. In summary, these results suggest that SPB ameliorates the inflammatory response induced by S. aureus LTA via suppressing the TLR2/NF-κB/NLRP3 signaling pathway, which indicates that SPB may be a potential agent for the treatment of bovine mastitis.
Collapse
Affiliation(s)
- Xin Wang
- College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang, Daqing 163319, China.
| | - Mengmeng Zhang
- College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang, Daqing 163319, China.
| | - Ning Jiang
- College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang, Daqing 163319, China.
| | - Aizhong Zhang
- College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang, Daqing 163319, China.
| |
Collapse
|
42
|
Kroon EE, Coussens AK, Kinnear C, Orlova M, Möller M, Seeger A, Wilkinson RJ, Hoal EG, Schurr E. Neutrophils: Innate Effectors of TB Resistance? Front Immunol 2018; 9:2637. [PMID: 30487797 PMCID: PMC6246713 DOI: 10.3389/fimmu.2018.02637] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/26/2018] [Indexed: 12/19/2022] Open
Abstract
Certain individuals are able to resist Mycobacterium tuberculosis infection despite persistent and intense exposure. These persons do not exhibit adaptive immune priming as measured by tuberculin skin test (TST) and interferon-γ (IFN-γ) release assay (IGRA) responses, nor do they develop active tuberculosis (TB). Genetic investigation of individuals who are able to resist M. tuberculosis infection shows there are likely a combination of genetic variants that contribute to the phenotype. The contribution of the innate immune system and the exact cells involved in this phenotype remain incompletely elucidated. Neutrophils are prominent candidates for possible involvement as primers for microbial clearance. Significant variability is observed in neutrophil gene expression and DNA methylation. Furthermore, inter-individual variability is seen between the mycobactericidal capacities of donor neutrophils. Clearance of M. tuberculosis infection is favored by the mycobactericidal activity of neutrophils, apoptosis, effective clearance of cells by macrophages, and resolution of inflammation. In this review we will discuss the different mechanisms neutrophils utilize to clear M. tuberculosis infection. We discuss the duality between neutrophils' ability to clear infection and how increasing numbers of neutrophils contribute to active TB severity and mortality. Further investigation into the potential role of neutrophils in innate immune-mediated M. tuberculosis infection resistance is warranted since it may reveal clinically important activities for prevention as well as vaccine and treatment development.
Collapse
Affiliation(s)
- Elouise E Kroon
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Anna K Coussens
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Infection and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Division of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Craig Kinnear
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Marianna Orlova
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,McGill International TB Centre, McGill University, Montreal, QC, Canada.,Departments of Medicine and Human Genetics, McGill University, Montreal, QC, Canada
| | - Marlo Möller
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Allison Seeger
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Robert J Wilkinson
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Medicine, Imperial College London, London, United Kingdom.,The Francis Crick Institute, London, United Kingdom
| | - Eileen G Hoal
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Erwin Schurr
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,McGill International TB Centre, McGill University, Montreal, QC, Canada.,Departments of Medicine and Human Genetics, McGill University, Montreal, QC, Canada
| |
Collapse
|
43
|
Wei R, Dhawan P, Baiocchi RA, Kim KY, Christakos S. PU.1 and epigenetic signals modulate 1,25-dihydroxyvitamin D 3 and C/EBPα regulation of the human cathelicidin antimicrobial peptide gene in lung epithelial cells. J Cell Physiol 2018; 234:10345-10359. [PMID: 30387140 DOI: 10.1002/jcp.27702] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/15/2018] [Indexed: 12/22/2022]
Abstract
LL-37, the only known human cathelicidin which is encoded by the human antimicrobial peptide (CAMP) gene, plays a critical role in protection against bacterial infection. We previously demonstrated that cathelicidin is induced by 1,25-dihydroxyvitamin D3 (1,25(OH) 2 D 3 ) in human airway epithelial cells with a resultant increase in bactericidal activity. In this study we identify key factors that co-operate with 1,25(OH) 2 D 3 in the regulation of CAMP. Our results show for the first time that PU.1, the myeloid transcription factor (which has also been identified in lung epithelial cells), co-operates with the vitamin D receptor and CCAAT/enhancer binding protein α (CEBPα) to enhance the induction of CAMP in lung epithelial cells. Our findings also indicate that enhancement of 1,25(OH) 2 D 3 regulation of CAMP by histone deacetylase inhibitors involves co-operation between acetylation and chromatin remodeling through Brahma-related gene 1 (BRG1; a component of the SWItch/sucrose nonfermentable [SWI/SNF] complex). BRG1 can be an activator or repressor depending on BRG1-associated factors. Protein arginine methyltransferase 5 (PRMT5), a methlytransferase which interacts with BRG1, represses 1,25(OH) 2 D 3 induced CAMP in part through dimethylation of H4R3. Our findings identify key mediators involved in the regulation of the CAMP gene in lung epithelial cells and suggest new approaches for therapeutic manipulation of gene expression to increase the antibacterial capability of the airway.
Collapse
Affiliation(s)
- Ran Wei
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, New Jersey
| | - Puneet Dhawan
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, New Jersey
| | - Robert A Baiocchi
- Department of Internal Medicine, Ohio State University, Columbus, Ohio
| | - Ki-Yoon Kim
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, New Jersey
| | - Sylvia Christakos
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, New Jersey
| |
Collapse
|
44
|
Simmons JD, Stein CM, Seshadri C, Campo M, Alter G, Fortune S, Schurr E, Wallis RS, Churchyard G, Mayanja-Kizza H, Boom WH, Hawn TR. Immunological mechanisms of human resistance to persistent Mycobacterium tuberculosis infection. Nat Rev Immunol 2018; 18:575-589. [PMID: 29895826 PMCID: PMC6278832 DOI: 10.1038/s41577-018-0025-3] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mycobacterium tuberculosis is a leading cause of mortality worldwide and establishes a long-lived latent infection in a substantial proportion of the human population. Multiple lines of evidence suggest that some individuals are resistant to latent M. tuberculosis infection despite long-term and intense exposure, and we term these individuals 'resisters'. In this Review, we discuss the epidemiological and genetic data that support the existence of resisters and propose criteria to optimally define and characterize the resister phenotype. We review recent insights into the immune mechanisms of M. tuberculosis clearance, including responses mediated by macrophages, T cells and B cells. Understanding the cellular mechanisms that underlie resistance to M. tuberculosis infection may reveal immune correlates of protection that could be utilized for improved diagnostics, vaccine development and novel host-directed therapeutic strategies.
Collapse
Affiliation(s)
- Jason D Simmons
- Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Catherine M Stein
- Department of Population & Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Chetan Seshadri
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Monica Campo
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Sarah Fortune
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| | - Erwin Schurr
- Program in Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - Robert S Wallis
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
- The Aurum Institute, Parktown, South Africa
| | | | | | - W Henry Boom
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Thomas R Hawn
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
45
|
Bekele A, Gebreselassie N, Ashenafi S, Kassa E, Aseffa G, Amogne W, Getachew M, Aseffa A, Worku A, Raqib R, Agerberth B, Hammar U, Bergman P, Aderaye G, Andersson J, Brighenti S. Daily adjunctive therapy with vitamin D 3 and phenylbutyrate supports clinical recovery from pulmonary tuberculosis: a randomized controlled trial in Ethiopia. J Intern Med 2018; 284:292-306. [PMID: 29696707 PMCID: PMC6202271 DOI: 10.1111/joim.12767] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Immunotherapy using vitamin D (vitD3 ) and phenylbutyrate (PBA) may support standard drug regimens used to treat infectious diseases. We investigated if vitD3 + PBA enhanced clinical recovery from pulmonary tuberculosis (TB). METHODS A randomized controlled trial was conducted in Addis Ababa, Ethiopia. Patients with smear-positive or smear-negative TB received daily oral supplementation with 5000 IU vitD3 and 2 × 500 mg PBA or placebo for 16 weeks, together with 6-month chemotherapy. Primary end-point: reduction of a clinical composite TB score at week 8 compared with baseline using modified intention-to-treat (mITT, n = 348) and per-protocol (n = 296) analyses. Secondary end-points: primary and modified TB scores (week 0, 4, 8, 16, 24), sputum conversion, radiological findings and plasma 25(OH)D3 concentrations. RESULTS Most subjects had low baseline plasma 25(OH)D3 levels that increased gradually in the vitD3 + PBA group compared with placebo (P < 0.0001) from week 0 to 16 (mean 34.7 vs. 127.4 nmol L-1 ). In the adjusted mITT analysis, the primary TB score was significantly reduced in the intervention group at week 8 (-0.52, 95% CI -0.93, -0.10; P = 0.015) while the modified TB score was reduced at week 8 (-0.58, 95% CI -1.02, -0.14; P = 0.01) and 16 (-0.34, 95% CI -0.64, -0.03; P = 0.03). VitD3 + PBA had no effect on longitudinal sputum-smear conversion (P = 0.98). Clinical adverse events were more common in the placebo group (24.3%) compared with the vitD3 + PBA group (12.6%). CONCLUSION Daily supplementation with vitD3 + PBA may ameliorate clinical TB symptoms and disease-specific complications, while the intervention had no effect on bacterial clearance in sputum.
Collapse
Affiliation(s)
- A Bekele
- Department of Internal Medicine, Faculty of Medicine, Black Lion University Hospital, Addis Ababa University, Addis Ababa, Ethiopia
| | - N Gebreselassie
- Center for Infectious Medicine (CIM), F59, Department of Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden.,Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - S Ashenafi
- Center for Infectious Medicine (CIM), F59, Department of Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - E Kassa
- Department of Internal Medicine, Faculty of Medicine, Black Lion University Hospital, Addis Ababa University, Addis Ababa, Ethiopia
| | - G Aseffa
- Department of Radiology, Faculty of Medicine, Black Lion University Hospital, Addis Ababa University, Addis Ababa, Ethiopia
| | - W Amogne
- Department of Internal Medicine, Faculty of Medicine, Black Lion University Hospital, Addis Ababa University, Addis Ababa, Ethiopia
| | - M Getachew
- Department of Internal Medicine, Faculty of Medicine, Black Lion University Hospital, Addis Ababa University, Addis Ababa, Ethiopia
| | - A Aseffa
- Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - A Worku
- Department of Public Health, Faculty of Medicine, Black Lion University Hospital, Addis Ababa University, Addis Ababa, Ethiopia
| | - R Raqib
- Infectious Diseases Division (IDD), International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - B Agerberth
- Clinical Microbiology, Department of Laboratory Medicine (Labmed), Karolinska Institutet, Stockholm, Sweden
| | - U Hammar
- Institute of Environmental Medicine (IMM), Karolinska Institutet, Stockholm, Sweden
| | - P Bergman
- Clinical Microbiology, Department of Laboratory Medicine (Labmed), Karolinska Institutet, Stockholm, Sweden
| | - G Aderaye
- Department of Internal Medicine, Faculty of Medicine, Black Lion University Hospital, Addis Ababa University, Addis Ababa, Ethiopia
| | - J Andersson
- Center for Infectious Medicine (CIM), F59, Department of Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden.,Division of Infectious Diseases, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - S Brighenti
- Center for Infectious Medicine (CIM), F59, Department of Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Abhimanyu, Coussens AK. The role of UV radiation and vitamin D in the seasonality and outcomes of infectious disease. Photochem Photobiol Sci 2018; 16:314-338. [PMID: 28078341 DOI: 10.1039/c6pp00355a] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The seasonality of infectious disease outbreaks suggests that environmental conditions have a significant effect on disease risk. One of the major environmental factors that can affect this is solar radiation, primarily acting through ultraviolet radiation (UVR), and its subsequent control of vitamin D production. Here we show how UVR and vitamin D, which are modified by latitude and season, can affect host and pathogen fitness and relate them to the outcomes of bacterial, viral and vector-borne infections. We conducted a thorough comparison of the molecular and cellular mechanisms of action of UVR and vitamin D on pathogen fitness and host immunity and related these to the effects observed in animal models and clinical trials to understand their independent and complementary effects on infectious disease outcome. UVR and vitamin D share common pathways of innate immune activation primarily via antimicrobial peptide production, and adaptive immune suppression. Whilst UVR can induce vitamin D-independent effects in the skin, such as the generation of photoproducts activating interferon signaling, vitamin D has a larger systemic effect due to its autocrine and paracrine modulation of cellular responses in a range of tissues. However, the seasonal patterns in infectious disease prevalence are not solely driven by variation in UVR and vitamin D levels across latitudes. Vector-borne pathogens show a strong seasonality of infection correlated to climatic conditions favoring their replication. Conversely, pathogens, such as influenza A virus, Mycobacterium tuberculosis and human immunodeficiency virus type 1, have strong evidence to support their interaction with vitamin D. Thus, UVR has both vitamin D-dependent and independent effects on infectious diseases; these effects vary depending on the pathogen of interest and the effects can be complementary or antagonistic.
Collapse
Affiliation(s)
- Abhimanyu
- Clinical Infectious Diseases Research Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Rd, Observatory, 7925, Western Cape, South Africa.
| | - Anna K Coussens
- Clinical Infectious Diseases Research Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Rd, Observatory, 7925, Western Cape, South Africa. and Division of Medical Microbiology, Department of Pathology, University of Cape Town, Anzio Rd, Observatory, 7925, Western Cape, South Africa
| |
Collapse
|
47
|
Abstract
Tuberculosis (TB) has troubled mankind for millennia, but current treatment strategies are long and complicated and the disease remains a major global health problem. The risk of Mycobacterium tuberculosis (Mtb) infection or progression of active TB disease is elevated in individuals with vitamin D deficiency. High-dose vitamin D was used to treat TB in the preantibiotic era, and in vitro experimental data show that vitamin D supports innate immune responses that restrict growth of Mtb. Several randomized controlled trials have tested whether adjunctive vitamin D supplementation enhances the clinical and microbiological response to standard antimicrobial chemotherapy for pulmonary TB. The effects have been modest at best, and attention is turning to the question of whether vitamin D supplementation might have a role in preventing acquisition or reactivation of latent Mtb infection. In this article, we describe the effects of vitamin D on host immune responses to Mtb in vitro and in vivo and review the results of clinical trials in the field. We also reflect on the findings of clinical trials of vitamin D supplementation for the prevention of acute respiratory tract infections, and discuss how these findings might influence the design of future trials to evaluate the role of vitamin D in the prevention and treatment of TB.
Collapse
Affiliation(s)
- S Brighenti
- Department of Medicine, Center for Infectious Medicine (CIM), Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - P Bergman
- Department of Laboratory Medicine (LABMED), Clinical Microbiology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - A R Martineau
- Blizard Institute, Centre for Immunobiology, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
48
|
DiNardo AR, Nishiguchi T, Mace EM, Rajapakshe K, Mtetwa G, Kay A, Maphalala G, Secor WE, Mejia R, Orange JS, Coarfa C, Bhalla KN, Graviss EA, Mandalakas AM, Makedonas G. Schistosomiasis Induces Persistent DNA Methylation and Tuberculosis-Specific Immune Changes. THE JOURNAL OF IMMUNOLOGY 2018; 201:124-133. [PMID: 29752313 DOI: 10.4049/jimmunol.1800101] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/09/2018] [Indexed: 12/29/2022]
Abstract
Epigenetic mechanisms, such as DNA methylation, determine immune cell phenotype. To understand the epigenetic alterations induced by helminth coinfections, we evaluated the longitudinal effect of ascariasis and schistosomiasis infection on CD4+ T cell DNA methylation and the downstream tuberculosis (TB)-specific and bacillus Calmette-Guérin-induced immune phenotype. All experiments were performed on human primary immune cells from a longitudinal cohort of recently TB-exposed children. Compared with age-matched uninfected controls, children with active Schistosoma haematobium and Ascaris lumbricoides infection had 751 differentially DNA-methylated genes, with 72% hypermethylated. Gene ontology pathway analysis identified inhibition of IFN-γ signaling, cellular proliferation, and the Th1 pathway. Targeted real-time quantitative PCR after methyl-specific endonuclease digestion confirmed DNA hypermethylation of the transcription factors BATF3, ID2, STAT5A, IRF5, PPARg, RUNX2, IRF4, and NFATC1 and cytokines or cytokine receptors IFNGR1, TNFS11, RELT (TNF receptor), IL12RB2, and IL12B (p < 0.001; Sidak-Bonferroni). Functional blockage of the IFN-γ signaling pathway was confirmed, with helminth-infected individuals having decreased upregulation of IFN-γ-inducible genes (Mann-Whitney p < 0.05). Hypomethylation of the IL-4 pathway and DNA hypermethylation of the Th1 pathway was confirmed by Ag-specific multidimensional flow cytometry demonstrating decreased TB-specific IFN-γ and TNF and increased IL-4 production by CD4+ T cells (Wilcoxon signed-rank p < 0.05). In S. haematobium-infected individuals, these DNA methylation and immune phenotypic changes persisted at least 6 mo after successful deworming. This work demonstrates that helminth infection induces DNA methylation and immune perturbations that inhibit TB-specific immune control and that the duration of these changes are helminth specific.
Collapse
Affiliation(s)
- Andrew R DiNardo
- The Global Tuberculosis Program, Immigrant and Global Health, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030;
| | - Tomoki Nishiguchi
- The Global Tuberculosis Program, Immigrant and Global Health, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030
| | - Emily M Mace
- Department of Pathology, Baylor College of Medicine, Houston, TX 77030.,Texas Children's Hospital Center for Human Immunobiology, Department of Pediatrics, Texas Children's Hospital Center for Human Immunobiology, Baylor College of Medicine, Houston, TX 77030
| | - Kimal Rajapakshe
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Godwin Mtetwa
- Baylor-Swaziland Children's Foundation, Mbabane H100, Swaziland
| | - Alexander Kay
- Baylor-Swaziland Children's Foundation, Mbabane H100, Swaziland
| | | | - W Evan Secor
- Parasitic Diseases Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA 30333
| | - Rojelio Mejia
- Department of Pediatrics, National School of Tropical Medicine, Texas Children's Hospital Center for Human Immunobiology, Houston, TX 77030
| | - Jordan S Orange
- Department of Pathology, Baylor College of Medicine, Houston, TX 77030.,Texas Children's Hospital Center for Human Immunobiology, Department of Pediatrics, Texas Children's Hospital Center for Human Immunobiology, Baylor College of Medicine, Houston, TX 77030
| | - Cristian Coarfa
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Kapil N Bhalla
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX 77030; and
| | - Edward A Graviss
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital Research Institute, Houston, TX 77030
| | - Anna M Mandalakas
- The Global Tuberculosis Program, Immigrant and Global Health, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030
| | - George Makedonas
- Department of Pathology, Baylor College of Medicine, Houston, TX 77030.,Texas Children's Hospital Center for Human Immunobiology, Department of Pediatrics, Texas Children's Hospital Center for Human Immunobiology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
49
|
Evaluation of the efficacy of valproic acid and suberoylanilide hydroxamic acid (vorinostat) in enhancing the effects of first-line tuberculosis drugs against intracellular Mycobacterium tuberculosis. Int J Infect Dis 2018; 69:78-84. [DOI: 10.1016/j.ijid.2018.02.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 01/29/2023] Open
|
50
|
Ramos-Espinosa O, Islas-Weinstein L, Peralta-Álvarez MP, López-Torres MO, Hernández-Pando R. The use of immunotherapy for the treatment of tuberculosis. Expert Rev Respir Med 2018; 12:427-440. [PMID: 29575946 DOI: 10.1080/17476348.2018.1457439] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Tuberculosis (TB) is the first cause of mortality by a single infectious agent in the world, causing more than one million deaths worldwide as reported by the World Health Organization (WHO). For the optimal control of TB infection, a protective immune response that limits bacterial spread without causing damage to the host is essential. Although most healthy individuals are capable of generating protective responses, patients who suffer pulmonary TB commonly present a defective immune function. Areas covered: We intend to highlight the potential of novel immunotherapeutic strategies that enhance and promote effective immune responses. The following methodology was undertaken for establishing a literature search: the authors used PubMed to search for 'Pulmonary Tuberculosis' and keywords that denoted the novel immunotherapeutic strategies discussed in length in the text including antibodies, antimicrobial peptides, cell therapy, cytokines and gene therapy. Expert commentary: The current therapeutic regimens for this disease are complex and involve the prolonged use of multiple antibiotics with diverse side effects that lead to therapeutic failure and bacterial resistance. The standard appliance of immunotherapy and its deployment to vulnerable populations will require coordinated work and may serve as a powerful tool to combat the ensuing threat of TB.
Collapse
Affiliation(s)
- Octavio Ramos-Espinosa
- a Section of Experimental Pathology, Department of Pathology , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , México
| | - León Islas-Weinstein
- a Section of Experimental Pathology, Department of Pathology , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , México
| | - Marco Polo Peralta-Álvarez
- a Section of Experimental Pathology, Department of Pathology , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , México.,b Laboratory of Immunochemistry, Department of Immunology , Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional , México City , México
| | - Manuel Othoniel López-Torres
- a Section of Experimental Pathology, Department of Pathology , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , México
| | - Rogelio Hernández-Pando
- a Section of Experimental Pathology, Department of Pathology , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , México
| |
Collapse
|