Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 173
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Front Immunol ; 12: 653571, 2021.
Article in English | MEDLINE | ID: mdl-34054815

ABSTRACT

Macrophages are indispensable immune cells tasked at eliminating intracellular pathogens. Mycobacterium tuberculosis (Mtb), one of the most virulent intracellular bacterial pathogens known to man, infects and resides within macrophages. While macrophages can be provoked by extracellular stimuli to inhibit and kill Mtb bacilli, these host defense mechanisms can be blocked by limiting nutritional metabolites, such as amino acids. The amino acid L-arginine has been well described to enhance immune function, especially in the context of driving macrophage nitric oxide (NO) production in mice. In this study, we aimed to establish the necessity of L-arginine on anti-Mtb macrophage function independent of NO. Utilizing an in vitro system, we identified that macrophages relied on NO for only half of their L-arginine-mediated host defenses and this L-arginine-mediated defense in the absence of NO was associated with enhanced macrophage numbers and viability. Additionally, we observed macrophage glycolysis to be driven by both L-arginine and mechanistic target of rapamycin (mTOR), and inhibition of glycolysis or mTOR reduced macrophage control of Mtb as well as macrophage number and viability in the presence of L-arginine. Our data underscore L-arginine as an essential nutrient for macrophage function, not only by fueling anti-mycobacterial NO production, but also as a central regulator of macrophage metabolism and additional host defense mechanisms.


Subject(s)
Arginine/metabolism , Dietary Supplements , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/diet therapy , Animals , Arginine/administration & dosage , Argininosuccinate Lyase/genetics , Argininosuccinate Lyase/metabolism , Argininosuccinate Synthase/genetics , Argininosuccinate Synthase/metabolism , Cell Survival , Disease Models, Animal , Humans , Macrophage Activation , Macrophages/metabolism , Mice , Mice, Knockout , Nitric Oxide/metabolism , Primary Cell Culture , RAW 264.7 Cells , Tuberculosis/immunology , Tuberculosis/microbiology
2.
J Infect Dev Ctries ; 15(3): 310-319, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33839703

ABSTRACT

Mycobacterium tuberculosis (M.tb) is the causative agent of tuberculosis (TB), an infectious disease that leads to numerous deaths worldwide. Malnutrition, smoking, alcohol abuse, Human Immunodeficiency Virus infection, and diabetes are some of the most important risk factors associated with TB development. At present, it is necessary to conduct studies on risk factors to establish new effective strategies and combat this disease. Malnutrition has been established as a risk factor since several years ago; although there is in vitro experimental evidence that reveals the importance of micronutrients in activating the immune response against M.tb, evidence from clinical trials is controversial. Currently, nutritional assessment is recommended in all TB patients upon diagnosis. However, there is insufficient evidence to indicate micronutrient supplementation as adjuvant therapy or prophylactic to prevent micronutrient depletion. Strengthening the interaction between basic and clinical research is necessary to carry out studies that will help establish adjuvant therapies to improve outcomes in TB patients. In this review, we discuss the experimental evidence, provided by basic research, regarding micronutrients in the TB field. However, when these studies are applied to clinical trials, the data are inconsistent, indicating that still missing mechanisms are necessary to propose alternatives to the treatment of TB patients.


Subject(s)
Malnutrition/complications , Micronutrients/immunology , Tuberculosis/etiology , Developing Countries/statistics & numerical data , Humans , Malnutrition/epidemiology , Malnutrition/prevention & control , Micronutrients/deficiency , Mycobacterium tuberculosis/immunology , Risk Factors , Tuberculosis/prevention & control
3.
Front Immunol ; 12: 623941, 2021.
Article in English | MEDLINE | ID: mdl-33777003

ABSTRACT

Host-directed therapies (HDTs) enhance the host response to tuberculosis (TB) infection to reduce disease severity. For instance, the manipulation of lipid mediator production diminishes the hyperactive immune response which is a known pathological feature of TB that generates lung tissue damage. Non-steroidal anti-inflammatory drugs (NSAIDs) and omega-3 long-chain polyunsaturated fatty acids (n-3 LCPUFA) are examples of such HDTs. In this mini-review, we recapitulate the literature available on the effects of NSAIDs and n-3 LCPUFA in TB as well as the immunological pathways underpinning these effects. Many NSAIDs have a great deal of data describing their effects and safety and in many jurisdictions are inexpensive, and sold over the counter in neighborhood convenience stores and supermarkets. The potential benefits of NSAIDs in TB are well-documented in pre-clinical studies. The reduction of pro-inflammatory lipid mediator production by inhibiting cyclooxygenase (COX) pathways with NSAIDs has been found to improve lung histopathology, bacterial control, and survival. Additionally, n-3 LCPUFA and its novel bioactive metabolites produced by COX and lipoxygenase (LOX) have been identified as safe and effective pro-resolving and antibacterial pharmaconutrients. Nevertheless, heterogeneous results have been reported in pre-clinical TB studies. Recently, the importance of the correct timing of NSAIDs and n-3 LCPUFA administration in TB has also been highlighted. This mini-review will provide a better understanding of the potential contribution of these therapies toward reducing inflammatory lung damage and improving bactericidal activity, especially during later stages of TB infection. It further highlights that clinical trials are required to confirm benefit and safety in TB patients.


Subject(s)
Antitubercular Agents/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Lipid Metabolism/drug effects , Lung/drug effects , Mycobacterium tuberculosis/drug effects , Tuberculosis, Pulmonary/drug therapy , Animals , Host-Pathogen Interactions , Humans , Lipoxygenase Inhibitors/therapeutic use , Lung/immunology , Lung/metabolism , Lung/microbiology , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Treatment Outcome , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/microbiology
4.
Biomed Pharmacother ; 138: 111489, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33743332

ABSTRACT

Guillain-Barré Syndrome (GBS), characterized by peripheral nerve demyelination and axonal damage, is initiated and aggravated through various of immunopathogenesis. Ginsenoside Rd, main active components extracted from ginseng saponins, is known to exhibit immune-regulate functions in many immune-mediated diseases. However, the evidence of preventive effect of Ginsenoside Rd on GBS is lacking. Experimental autoimmune neuritis (EAN) mice, classic model of GBS, were established and treated with GSRd or vehicle. Clinical score and nerve tissue histomorphology were evaluated. Monocytes in peripheral blood and tissue were detected by flow cytometry analysis and immunofluorescence staining. For the in vitro study, GSRd and vehicle were added in the culture medium to assess their regulatory function on monocytes phenotype. In vivo data showed a protective role of GSRd on alleviating symptoms and tissue damage on Day 20 and 25. Administration of GSRd increased non-classical Ly6Clo monocytes in both peripheral blood and injured nerve tissue, and also switched tissue macrophages phenotype into resolution-phase. In vitro study indicated similar role of GSRd on monocytes differentiation status. Transcription factors like Nr4a1 were elevated after GSRd treatment. These findings revealed the protective role of GSRd against EAN, and potential preventive function on GBS patients.


Subject(s)
Ginsenosides/therapeutic use , Monocytes/drug effects , Monocytes/immunology , Neuritis, Autoimmune, Experimental/drug therapy , Neuritis, Autoimmune, Experimental/immunology , Panax , Amino Acid Sequence , Animals , Dose-Response Relationship, Drug , Ginsenosides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Sciatic Neuropathy/drug therapy , Sciatic Neuropathy/immunology
5.
Tuberculosis (Edinb) ; 127: 102045, 2021 03.
Article in English | MEDLINE | ID: mdl-33434785

ABSTRACT

HIV infection is a major risk factor predisposing for Mycobacterium tuberculosis infection and progression to active tuberculosis (TB). As host immune response defines the course of infection, we aimed to identify immuno-endocrine changes over six-months of anti-TB chemotherapy in HIV+ people. Plasma levels of cortisol, DHEA and DHEA-S, percentages of CD4+ regulatory T cell subsets and number of IFN-γ-secreting cells were determined. Several cytokines, chemokines and C-reactive protein levels were measured. Results were correlated with clinical parameters as predictors of infection resolution and compared to similar data from HIV+ individuals, HIV-infected persons with latent TB infection and healthy donors. Throughout the course of anti-TB/HIV treatment, DHEA and DHEA-S plasma levels raised while cortisol diminished, which correlated to predictive factors of infection resolution. Furthermore, the balance between cortisol and DHEA, together with clinical assessment, may be considered as an indicator of clinical outcome after anti-TB treatment in HIV+ individuals. Clinical improvement was associated with reduced frequency of unconventional Tregs, increment in IFN-γ-secreting cells, diminution of systemic inflammation and changes of circulating cytokines and chemokines. This study suggests that the combined anti-HIV/TB therapies result in partial restoration of both, immune function and adrenal hormone plasma levels.


Subject(s)
Adrenal Cortex Hormones/blood , Antitubercular Agents/therapeutic use , HIV Infections/blood , HIV-1/pathogenicity , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Adult , Biomarkers/blood , Coinfection , Cytokines/blood , Dehydroepiandrosterone/blood , Dehydroepiandrosterone Sulfate/blood , Female , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Host-Pathogen Interactions , Humans , Hydrocortisone/blood , Male , Middle Aged , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Prospective Studies , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/microbiology , T-Lymphocytes, Regulatory/virology , Time Factors , Treatment Outcome , Tuberculosis/blood , Tuberculosis/immunology , Tuberculosis/microbiology
6.
Mucosal Immunol ; 14(1): 253-266, 2021 01.
Article in English | MEDLINE | ID: mdl-32862202

ABSTRACT

Mycobacterium tuberculosis (Mtb) infection induces pulmonary expression of the heme-degrading enzyme heme oxygenase-1 (HO-1). We have previously shown that pharmacological inhibition of HO-1 activity in experimental tuberculosis results in decreased bacterial loads and unexpectedly that this outcome depends on the presence of T lymphocytes. Here, we extend these findings by demonstrating that IFNγ production by T lymphocytes and NOS2 expression underlie this T-cell requirement and that HO-1 inhibition potentiates IFNγ-induced NOS2-dependent control of Mtb by macrophages in vitro. Among the products of heme degradation by HO-1 (biliverdin, carbon monoxide, and iron), only iron supplementation reverted the HO-1 inhibition-induced enhancement of bacterial control and this reversal was associated with decreased NOS2 expression and NO production. In addition, we found that HO-1 inhibition results in decreased labile iron levels in Mtb-infected macrophages in vitro and diminished iron accumulation in Mtb-infected lungs in vivo. Together these results suggest that the T-lymphocyte dependence of the therapeutic outcome of HO-1 inhibition on Mtb infection reflects the role of the enzyme in generating iron that suppresses T-cell-mediated IFNγ/NOS2-dependent bacterial control. In broader terms, our findings highlight the importance of the crosstalk between iron metabolism and adaptive immunity in determining the outcome of infection.


Subject(s)
Heme Oxygenase-1/antagonists & inhibitors , Host-Pathogen Interactions , Interferon-gamma/metabolism , Mycobacterium tuberculosis , Nitric Oxide Synthase Type II/metabolism , Tuberculosis/metabolism , Tuberculosis/microbiology , Animals , Bacterial Load , Host-Pathogen Interactions/immunology , Iron/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Models, Biological , Mycobacterium tuberculosis/immunology , Nitric Oxide/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tuberculosis/immunology
7.
J Allergy Clin Immunol ; 146(3): 652-666.e11, 2020 09.
Article in English | MEDLINE | ID: mdl-32445838

ABSTRACT

BACKGROUND: Effector functions of IgG Abs are regulated by their Fc N-glycosylation pattern. IgG Fc glycans that lack galactose and terminal sialic acid residues correlate with the severity of inflammatory (auto)immune disorders and have also been linked to protection against viral infection and discussed in the context of vaccine-induced protection. In contrast, sialylated IgG Abs have shown immunosuppressive effects. OBJECTIVE: We sought to investigate IgG glycosylation programming during the germinal center (GC) reaction following immunization of mice with a foreign protein antigen and different adjuvants. METHODS: Mice were analyzed for GC T-cell, B-cell, and plasma cell responses, as well as for antigen-specific serum IgG subclass titers and Fc glycosylation patterns. RESULTS: Different adjuvants induce distinct IgG+ GC B-cell responses with specific transcriptomes and expression levels of the α2,6-sialyltransferase responsible for IgG sialylation that correspond to distinct serum IgG Fc glycosylation patterns. Low IgG Fc sialylation programming in GC B cells was overall highly dependent on the Foxp3- follicular helper T (TFH) cell-inducing cytokine IL-6, here in particular induced by water-in-oil adjuvants and Mycobacterium tuberculosis. Furthermore, low IgG Fc sialylation programming was dependent on adjuvants that induced IL-27 receptor-dependent IFN-γ+ TFH1 cells, IL-6/IL-23-dependent IL-17A+ TFH17 cells, and high ratios of TFH cells to Foxp3+ follicular regulatory T cells. Here, the 2 latter were dependent on M tuberculosis and its cord factor. CONCLUSION: This study's findings regarding adjuvant-dependent GC responses and IgG glycosylation programming may aid in the development of novel vaccination strategies to induce IgG Abs with both high affinity and defined Fc glycosylation patterns in the GC.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens/administration & dosage , Germinal Center/immunology , Immunoglobulin G/immunology , Alum Compounds/administration & dosage , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cytokines/immunology , Female , Freund's Adjuvant/administration & dosage , Glycosylation , Lipopolysaccharides/administration & dosage , Mice, Inbred C57BL , Mice, Knockout , Mineral Oil/administration & dosage , Mycobacterium tuberculosis/immunology , Ovalbumin/administration & dosage , Polysorbates/administration & dosage , Squalene/administration & dosage , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Vaccination
8.
Tuberculosis (Edinb) ; 120: 101902, 2020 01.
Article in English | MEDLINE | ID: mdl-32090863

ABSTRACT

Despite the availability of potent antitubercular drugs, tuberculosis (TB) still remains one of the world's leading causes of death. The current antitubercular therapy (ATT) suffers from a drawback of longer duration that imposes a major challenge of patient non compliance and resistance development. The current scenario necessitates alternative strategies with potential to shorten treatment duration that could pave the way for improved clinical outcomes. In recent years, host directed adjunctive therapies have raised considerable attention and emerged as a promising intervention which targets clinically relevant biological pathways in hosts to modulate pathological immune responses. Few of the approved drugs namely statins, metformin, ibuprofen, aspirin, valproic acid, adalimumab, bevacizumab, zileuton and vitamin D3 have shown promising results in clinical outcomes during their preliminary screening in TB patients and can be potentially repurposed as antitubercular drugs. This review highlights clinical and non clinical evidences of some already existing drug and their targets in hosts that could help in shortening treatment duration and reducing bacterial burden at minimal doses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antitubercular Agents/administration & dosage , Drug Repositioning , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Adjuvants, Immunologic/adverse effects , Administration, Oral , Antitubercular Agents/adverse effects , Drug Resistance, Bacterial , Drug Therapy, Combination , Humans , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Treatment Outcome , Tuberculosis/immunology , Tuberculosis/microbiology
9.
J Biomed Sci ; 27(1): 20, 2020 Jan 06.
Article in English | MEDLINE | ID: mdl-31906962

ABSTRACT

BACKGROUND: Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis (TB), affecting approximately one third of the world's population. Development of an adequate immune response will determine disease progression or progress to chronic infection. Risk of developing TB among human immunodeficiency virus (HIV)-coinfected patients (HIV-TB) is 20-30 times higher than those without HIV infection, and a synergistic interplay between these two pathogens accelerates the decline in immunological functions. TB treatment in HIV-TB coinfected persons is challenging and it has a prolonged duration, mainly due to the immune system failure to provide an adequate support for the therapy. Therefore, we aimed to study the role of the hormone 7-oxo-dehydroepiandrosterone (7-OD) as a modulator of anti-tuberculosis immune responses in the context of HIV-TB coinfection. METHODS: A cross-sectional study was conducted among HIV-TB patients and healthy donors (HD). We characterized the ex vivo phenotype of CD4 + T cells and also evaluated in vitro antigen-specific responses by Mtb stimulation of peripheral blood mononuclear cells (PBMCs) in the presence or absence of 7-OD. We assessed lymphoproliferative activity, cytokine production and master transcription factor profiles. RESULTS: Our results show that HIV-TB patients were not able to generate successful anti-tubercular responses in vitro compared to HD, as reduced IFN-γ/IL-10 and IFN-γ/IL-17A ratios were observed. Interestingly, treatment with 7-OD enhanced Th1 responses by increasing Mtb-induced proliferation and the production of IFN-γ and TNF-α over IL-10 levels. Additionally, in vitro Mtb stimulation augmented the frequency of cells with a regulatory phenotype, while 7-OD reduced the proportion of these subsets and induced an increase in CD4 + T-bet+ (Th1) subpopulation, which is associated with clinical data linked to an improved disease outcome. CONCLUSIONS: We conclude that 7-OD modifies the cytokine balance and the phenotype of CD4 + T cells towards a more favorable profile for mycobacteria control. These results provide new data to delineate novel treatment approaches as co-adjuvant for the treatment of TB.


Subject(s)
Coinfection/immunology , Dehydroepiandrosterone/analogs & derivatives , HIV Infections/immunology , HIV-1/immunology , Mycobacterium tuberculosis/immunology , Th1 Cells/immunology , Tuberculosis, Pulmonary/immunology , Adult , Chronic Disease , Coinfection/pathology , Cross-Sectional Studies , Dehydroepiandrosterone/immunology , Dehydroepiandrosterone/pharmacology , Female , HIV Infections/pathology , Humans , Male , Middle Aged , Th1 Cells/pathology , Tuberculosis, Pulmonary/pathology
10.
Vet Immunol Immunopathol ; 215: 109884, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31420066

ABSTRACT

Many vaccines against childhood diseases are administered early after birth, but vaccine development studies frequently test efficacy in adult rather than in neonatal animal models. In countries with endemic tuberculosis (TB), Bacillus Calmette-Guerin (BCG) is administered as part of the neonatal vaccine regimen because it prevents against the disseminated form of TB in children, although it has variable efficacy against pulmonary TB. Several promising new vaccines against TB are currently being tested in adult animal models. Here we evaluated neonatal piglets as an animal model to test vaccine efficacy. For this purpose, minipigs were vaccinated or not with BCG 48 h after birth and their immune response followed longitudinally until adolescence. We characterized the memory and activation phenotype of T cells, cytokine profile, and monocyte activation in response to BCG stimulation from 4 weeks of age into adolescence- age of 24 weeks. Immunological responses in vaccinated and non-vaccinated animals were further monitored upon infection with a low dose exposure to Mycobacterium tuberculosis strain HN878 via the aerosol route. Comparing the immunological response elicited by BCG vaccination in minipigs vs similar studies in infants, suggest that minipigs have the potential to serve as an effective neonatal animal model for vaccine development.


Subject(s)
BCG Vaccine/immunology , Disease Models, Animal , Mycobacterium tuberculosis/immunology , Swine, Miniature/immunology , Tuberculosis, Pulmonary/immunology , Animals , Animals, Newborn , Cytokines/metabolism , Drug Evaluation, Preclinical/methods , Female , Immunogenicity, Vaccine , Immunologic Memory , Immunophenotyping , Longitudinal Studies , Lymphocyte Activation , Male , Monocytes/immunology , Swine , Tuberculosis/immunology , Tuberculosis, Pulmonary/prevention & control
11.
Front Immunol ; 10: 1349, 2019.
Article in English | MEDLINE | ID: mdl-31293568

ABSTRACT

Liposomes have been long considered as a vaccine delivery system but this technology remains to be fully utilized. Here, we describe a novel liposome-based subunit vaccine formulation for tuberculosis (TB) based on phosphatidylserine encapsulating two prominent TB antigens, Ag85B, and ESAT-6. We show that the resulting liposomes (Lipo-AE) are stable upon storage and can be readily taken up by antigen presenting cells and that their antigenic cargo is delivered and processed within endosomal cell compartments. The Lipo-AE vaccine formulation combined with the PolyIC adjuvant induced a mixed Th1/Th17-Th2 immune response to Ag85B but only a weak response to ESAT-6. An immunization regimen based on systemic delivery followed by mucosal boost with Lipo-AE resulted in the accumulation of resident memory T cells in the lungs. Most importantly though, when Lipo-AE vaccine candidate was administered to BCG-immunized mice subsequently challenged with low dose aerosol Mycobacterium tuberculosis, we observed a significant reduction of the bacterial load in the lungs and spleen compared to BCG alone. We therefore conclude that the immunization with mycobacterial antigens delivered by phosphatidylserine based liposomes in combination with Poly:IC adjuvant may represent a novel BCG boosting vaccination strategy.


Subject(s)
Acyltransferases/immunology , Antigens, Bacterial/immunology , BCG Vaccine/immunology , Bacterial Proteins/immunology , Liposomes/immunology , Tuberculosis, Pulmonary/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Bacterial Load , Drug Delivery Systems , Female , Humans , Immunologic Memory/immunology , Lung/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Phosphatidylserines/immunology , Poly I-C/immunology , Spleen/microbiology , T-Lymphocytes, Helper-Inducer/immunology , Vaccination , Vaccines, Subunit/immunology
12.
World J Gastroenterol ; 25(26): 3291-3298, 2019 Jul 14.
Article in English | MEDLINE | ID: mdl-31341356

ABSTRACT

Solid organ transplantation (SOT) is the best treatment option for end-stage organ disease. Newer immunosuppressive agents have reduced the incidence of graft rejection but have increased the risk of infection, particularly due to the reactivation of latent infections due to opportunistic agents such as Mycobacterium tuberculosis. Active tuberculosis (TB) after SOT is a significant cause of morbidity and mortality. Most cases of posttransplant TB are secondary to reactivation of latent tuberculosis infection (LTBI) due to the effects of long-term immunosuppressive therapy. Risk minimization strategies have been developed to diagnose LTBI and initiate treatment prior to transplantation. Isoniazid with vitamin B6 supplementation is the treatment of choice. However, liver transplantation (LT) candidates and recipients have an increased risk of isoniazid-induced liver toxicity, leading to lower treatment completion rates than in other SOT populations. Fluoroquinolones (FQs) exhibit good in vitro antimycobacterial activity and a lower risk of drug-induced liver injury than isoniazid. In the present review, we highlight the disease burden posed by posttransplant TB and summarize the emerging clinical evidence supporting the use of FQs for the treatment of LTBI in LT recipients and candidates.


Subject(s)
Antitubercular Agents/administration & dosage , Fluoroquinolones/administration & dosage , Latent Tuberculosis/drug therapy , Liver Transplantation/adverse effects , Postoperative Complications/drug therapy , Allografts , Antitubercular Agents/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/prevention & control , End Stage Liver Disease/surgery , Graft Rejection/epidemiology , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Immunosuppressive Agents/adverse effects , Incidence , Isoniazid/adverse effects , Latent Tuberculosis/immunology , Latent Tuberculosis/microbiology , Liver , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/isolation & purification , Postoperative Complications/immunology , Postoperative Complications/microbiology , Risk Factors , Treatment Outcome
13.
Front Immunol ; 10: 934, 2019.
Article in English | MEDLINE | ID: mdl-31130946

ABSTRACT

Most microbes invading through mucosal surfaces cause disease and therefore strategies to induce mucosal immune responses are strongly needed. Vitamin A metabolites, such as retinoic acid (RA), play crucial roles in programming T and B cells to home to mucosal compartments, therefore we evaluated the capacity of RA to elicit mucosal immune responses against tuberculosis (TB) after parenteral vaccination. We found that mice immunized through subcutaneous injections with the TB subunit vaccine (CAF01+H56) in presence of RA show enhanced mucosal H56-specific IgA responses and enhanced Ag-specific CD4+ T lymphocytes homing to the lung as compared with control mice. Immunization with CAF01+H56 in presence of RA resulted in lower bacterial loads in the lungs of mice 14 days after challenge with virulent Mycobacterium tuberculosis (Mtb) as compared to mice immunized in the absence of RA or vaccinated with BCG. Higher amounts of IFNγ and IL-17 pro-inflammatory cytokines were found in lung homogenates of mice immunized with CAF01+H56 and RA 24 h after Mtb infection. However, 6 weeks after infection the protection was comparable in vaccinated mice with or without RA even though treatment with RA during immunization is able to better contain the inflammatory response by the host. Furthermore, at later stage of the infection a higher percentage of Mtb specific CD4+PD1+ T lymphocytes were found in the lungs of mice immunized with CAF01+H56 and RA. These data show that an enhanced mucosal immune response is generated during parenteral vaccination in presence of RA. Furthermore, RA treatment contained the bacterial growth at an early stage of the infection and limited the inflammatory response in the lung at later time points.


Subject(s)
Immunity, Mucosal , Tretinoin/administration & dosage , Tuberculosis Vaccines/administration & dosage , Tuberculosis, Pulmonary/prevention & control , Vaccines, Subunit/administration & dosage , Allergens/immunology , Animals , Antibodies, Bacterial/immunology , Antibody-Producing Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Female , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Lung/immunology , Lymphocytes/immunology , Mice , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/immunology , Ovalbumin/immunology , Tuberculosis, Pulmonary/immunology , Vaccination
14.
Front Immunol ; 10: 206, 2019.
Article in English | MEDLINE | ID: mdl-30853958

ABSTRACT

Mucosal associated invariant T (MAIT) cells are unconventional, semi-invariant T lymphocytes that recognize microbial-derived vitamin B2 (riboflavin) biosynthesis precursor derivatives presented by the monomorphic MHC class 1-related (MR1) molecule. Upon microbial infection, MAIT cells rapidly produce cytokines and cytotoxic effectors, and are thus important players in anti-microbial defense. MAIT cells are protective in experimental models of infection and are decreased in the blood of adult patients with bacterial infections, including Mycobacterium tuberculosis (Mtb). In children, the risk of rapid progression to active tuberculosis (TB) following Mtb infection is higher than in adults. Whether MAIT cells influence the outcome of Mtb infection in children is therefore, an important issue. We analyzed MAIT cell numbers and phenotype in 115 children investigated for pulmonary TB and determined their potential correlation with disease progression. MAIT cells were reduced in numbers and activated in the peripheral blood of children with active TB as compared to those with latent TB infection (LTBI) and healthy children. Moreover, MAIT cells did not accumulate and did not proliferate in the lung of children with active TB. These results suggest that MAIT cells may be important in preventing progression of Mtb infection to active TB in children.


Subject(s)
Lymphocyte Count , Mucosal-Associated Invariant T Cells/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Adolescent , Age Factors , Biomarkers , Child , Child, Preschool , Histocompatibility Antigens Class I/immunology , Humans , Immunity, Innate , Immunity, Mucosal , Immunophenotyping , Infant , Lung/immunology , Lung/microbiology , Lung/pathology , Mucosal-Associated Invariant T Cells/metabolism , Tuberculosis, Pulmonary/blood , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/metabolism
15.
Am J Clin Dermatol ; 20(4): 483-491, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30919314

ABSTRACT

BACKGROUND: Biologic therapies pose a risk for opportunistic infections, especially for reactivating latent tuberculosis infection (LTBI). OBJECTIVE: The aim was to describe the clinical features and mortality rate of active Mycobacterium tuberculosis (TB) in psoriasis patients receiving biologic therapies. METHODS: A systematic review of PubMed, Google Scholar, ScienceDirect, Cochrane Library, and ClinicalTrials.gov databases was performed. Studies describing active TB in patients with psoriasis receiving biologic therapy from inception to May 31, 2018 were included. Clinical data as well as mortality rates were recorded. RESULTS: Fifty-one studies were included, evaluating 78 patients with active TB: 11 prospective studies, 13 retrospective, and 27 case reports/series. Most patients (73%) with active TB were male, the mean age was 48 ± 13 years, and 85% were of European or Asian origin. Pre-treatment LTBI screening was negative for 63% of patients. Disease presented in 33% of patients within the first 3 months of treatment, and in 51% within the first 6 months. Most patients (72%) presented with extra-pulmonary TB, and 49% had disseminated disease. The mortality rate was 7%. LIMITATIONS: Limitations of this review are its small sample size and inclusion of case reports. CONCLUSIONS: Some patients develop active TB despite LTBI screening. Clinicians initiating biologic therapy in patients with psoriasis should be aware of the clinical features of active TB in this scenario.


Subject(s)
Biological Therapy/adverse effects , Opportunistic Infections/complications , Psoriasis/drug therapy , Tuberculosis/complications , Adalimumab/adverse effects , Etanercept/adverse effects , Humans , Immunocompromised Host , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/isolation & purification , Opportunistic Infections/immunology , Opportunistic Infections/mortality , Psoriasis/complications , Psoriasis/immunology , Tuberculosis/immunology , Tuberculosis/mortality
16.
Invest Ophthalmol Vis Sci ; 60(1): 420-429, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30695094

ABSTRACT

Purpose: We determine the changes in intestinal microbiota and/or disruptions in intestinal homeostasis during uveitis. Methods: Experimental autoimmune uveitis (EAU) was induced in B10.RIII mice with coadministration of interphotoreceptor retinoid-binding protein peptide (IRBP) and killed mycobacterial antigen (MTB) as an adjuvant. Using 16S rRNA gene sequencing, we looked at intestinal microbial differences during the course of uveitis, as well as intestinal morphologic changes, changes in intestinal permeability by FITC-dextran leakage, antimicrobial peptide expression in the gastrointstinal tract, and T lymphocyte prevalence before and at peak intraocular inflammation. Results: We demonstrate that increased intestinal permeability and antimicrobial peptide expression in the intestinal tract coincide in timing with increased effector T cells in the mesenteric lymph nodes, during the early stages of uveitis, before peak inflammation. Morphologic changes in the intestine were most prominent during this phase, but also occurred with adjuvant MTB alone, whereas increased intestinal permeability was found only in IRBP-immunized mice that develop uveitis. We also demonstrate that the intestinal microbiota were altered during the course of uveitis, and that some of these changes are specific to uveitic animals, whereas others are influenced by adjuvant MTB alone. Intestinal permeability peaked at 2 weeks, coincident with an increase in intestinal bacterial strain differences, peak lipocalin production, and peak uveitis. Conclusions: An intestinal dysbiosis accompanies a disruption in intestinal homeostasis in autoimmune uveitis, although adjuvant MTB alone promotes intestinal disruption as well. This may indicate a novel axis for future therapeutic targeting experimentally or clinically.


Subject(s)
Autoimmune Diseases/microbiology , Dysbiosis/microbiology , Gastrointestinal Microbiome/immunology , Homeostasis/physiology , Intestines/physiology , Uveitis/microbiology , Animals , Antigens, Bacterial/immunology , Autoimmune Diseases/immunology , Enzyme-Linked Immunosorbent Assay , Eye Proteins , Flow Cytometry , Lipocalins/metabolism , Mice , Mice, Mutant Strains , Models, Animal , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , RNA, Ribosomal, 16S/genetics , Retinol-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/immunology , Uveitis/immunology , Zonula Occludens-1 Protein/metabolism
17.
J Infect Dis ; 219(9): 1356-1363, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30476125

ABSTRACT

Almost 800 million people are chronically undernourished worldwide, of whom 98% are in low- and middle-income countries where tuberculosis is endemic. In many tuberculosis-endemic countries, undernutrition is a driver of tuberculosis incidence and associated with a high population attributable fraction of tuberculosis and poor treatment outcomes. Data suggest that undernutrition impairs innate and adaptive immune responses needed to control Mycobacterium tuberculosis infection and may affect responses to live vaccines, such as BCG. Given its impact on tuberculosis, addressing undernutrition will be a vital component of the World Health Organization End TB strategy. This narrative review describes the effect of undernutrition on the immune response, vaccine response, and tuberculosis incidence, severity, and treatment outcomes.


Subject(s)
Malnutrition/epidemiology , Malnutrition/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/epidemiology , Comorbidity , Dietary Supplements , Humans , Incidence , Nutrients/therapeutic use , Nutrition Assessment , Public Health , Severity of Illness Index , Treatment Outcome , Vaccines/immunology
18.
Front Immunol ; 9: 2438, 2018.
Article in English | MEDLINE | ID: mdl-30410488

ABSTRACT

Identifying a blood circulating cellular biomarker that can be used to assess severity of disease and predict the time to culture conversion (TCC) in patients with multidrug resistant tuberculosis (MDR-TB) would facilitate monitoring response to treatment and may be of value in the design of future drug trials. We report on the frequency of blood Ki67+HLA-DR- CD4+ T regulatory (Treg) cells in predicting microbiological outcome before initiating second-line treatment for MDR-TB. Fifty-one patients with MDR-TB were enrolled and followed over 18 months; a subset of patients was sputum culture (SC) negative at baseline (n = 9). SC positive patients were divided into two groups, based on median TCC: rapid responders (≤71 days TCC; n = 21) and slow responders (>71 days TCC; n = 21). Whole blood at baseline, months 2 and 6 was stimulated with M tuberculosis (Mtb) antigens and Treg cells were then identified as CD3+CD4+CD25hiFoxP3+CD127-CD69- and further delineated as Ki67+HLA-DR- Treg. The frequency of these cells was significantly enlarged at baseline in SC positive relative to SC negative and smear positive relative to smear negative patients and in those with lung cavitation. This difference was further supported by unsupervised hierarchical clustering showing a significant grouping at baseline of total and early differentiated memory Treg cells in slow responders. Conversely, there was a clustering of a lower proportion of Treg cells and activated IFNγ-expressing T cells at baseline in the rapid responders. Examining changes over time revealed a more gradual reduction of Treg cells in slow responders relative to rapid responders to treatment. Receiver operating curve analysis showed that baseline Mtb-stimulated Ki67+HLA-DR- Treg cells could predict the TCC of MDR-TB treatment response with 81.2% sensitivity and 85% specificity (AUC of 0.87, p < 0.0001), but this was not the case after 2 months of treatment. In conclusion, our data show that the frequency of a highly defined Mtb-stimulated blood Treg cell population at baseline can discriminate MDR-TB disease severity and predict time to culture clearance.


Subject(s)
Lymphocyte Count , Mycobacterium tuberculosis/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tuberculosis, Multidrug-Resistant/immunology , Tuberculosis, Multidrug-Resistant/metabolism , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Female , HLA-DR Antigens/metabolism , Humans , Immunologic Memory , Ki-67 Antigen/metabolism , Male , Microbial Sensitivity Tests , ROC Curve , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tuberculosis, Multidrug-Resistant/diagnosis , Tuberculosis, Multidrug-Resistant/drug therapy
19.
Front Immunol ; 9: 1778, 2018.
Article in English | MEDLINE | ID: mdl-30166982

ABSTRACT

It is known that vitamin B1 (VB1) has a protective effect against oxidative retinal damage induced by anti-tuberculosis drugs. However, it remains unclear whether VB1 regulates immune responses during Mycobacterium tuberculosis (MTB) infection. We report here that VB1 promotes the protective immune response to limit the survival of MTB within macrophages and in vivo through regulation of peroxisome proliferator-activated receptor γ (PPAR-γ). VB1 promotes macrophage polarization into classically activated phenotypes with strong microbicidal activity and enhanced tumor necrosis factor-α and interleukin-6 expression at least in part by promoting nuclear factor-κB signaling. In addition, VB1 increases mitochondrial respiration and lipid metabolism and PPAR-γ integrates the metabolic and inflammatory signals regulated by VB1. Using both PPAR-γ agonists and deficient mice, we demonstrate that VB1 enhances anti-MTB activities in macrophages and in vivo by down-regulating PPAR-γ activity. Our data demonstrate important functions of VB1 in regulating innate immune responses against MTB and reveal novel mechanisms by which VB1 exerts its function in macrophages.


Subject(s)
Immunity, Innate , Mycobacterium tuberculosis/immunology , PPAR gamma/metabolism , Thiamine/metabolism , Tuberculosis/immunology , Tuberculosis/metabolism , Animals , Biomarkers , Host-Pathogen Interactions/immunology , Immunity, Innate/drug effects , Immunophenotyping , Lipid Metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Transgenic , Mitochondria/metabolism , NF-kappa B/metabolism , Signal Transduction , Thiamine/pharmacology , Tuberculosis/microbiology
20.
Front Immunol ; 9: 2069, 2018.
Article in English | MEDLINE | ID: mdl-30258443

ABSTRACT

Mycobacterium tuberculosis (M. tb), the causative bacterial agent responsible for tuberculosis (TB) continues to afflict millions of people worldwide. Although the human immune system plays a critical role in containing M. tb infection, elimination proves immensely more challenging. Consequently, there has been a worldwide effort to eradicate, and limit the spread of M. tb through the conventional use of first-line antibiotics. Unfortunately, with the emergence of drug resistant and multi-drug resistant strains of M. tb the archetypical antibiotics no longer provide the same ascendancy as they once did. Furthermore, when administered, these first-line antibiotics commonly present severe complications and side effects. The biological antioxidant glutathione (GSH) however, has been demonstrated to have a profound mycobactericidal effect with no reported adverse consequences. Therefore, we examined if N-Acetyl Cysteine (NAC), the molecular precursor to GSH, when supplemented in combination with suboptimal levels of standalone first-line antibiotics would be sufficient to completely clear M. tb infection within in vitro derived granulomas from healthy subjects and individuals with type 2 diabetes (T2DM). Our results revealed that by virtue of immune modulation, the addition of NAC to subprime levels of isoniazid (INH) and rifampicin (RIF) was indeed capable of inducing complete clearance of M. tb among healthy individuals.


Subject(s)
Acetylcysteine/administration & dosage , Anti-Bacterial Agents/administration & dosage , Granuloma, Respiratory Tract , Mycobacterium tuberculosis/immunology , Tuberculosis, Multidrug-Resistant , Tuberculosis, Pulmonary , Adult , Aged , Female , Granuloma, Respiratory Tract/drug therapy , Granuloma, Respiratory Tract/immunology , Granuloma, Respiratory Tract/pathology , Humans , Male , Middle Aged , Tuberculosis, Multidrug-Resistant/drug therapy , Tuberculosis, Multidrug-Resistant/immunology , Tuberculosis, Multidrug-Resistant/pathology , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology
SELECTION OF CITATIONS
SEARCH DETAIL