151
|
Elkington PT, Friedland JS. Permutations of time and place in tuberculosis. THE LANCET. INFECTIOUS DISEASES 2015; 15:1357-60. [PMID: 26321650 PMCID: PMC4872044 DOI: 10.1016/s1473-3099(15)00135-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 06/04/2015] [Accepted: 06/09/2015] [Indexed: 02/01/2023]
Abstract
Tuberculosis remains a global health pandemic. The current depiction of the Mycobacterium tuberculosis life cycle proposes that airborne bacilli are inhaled and phagocytosed by alveolar macrophages, resulting in the formation of a granuloma that ruptures into the airways to reinitiate the infectious cycle. However, this widely proposed model overlooks the fact, established 100 years ago, that the initial site of M tuberculosis implantation is in the lower zones of the lungs, whereas infectious cavitary pulmonary disease develops at the lung apices. The immunological events at these two pulmonary locations are different--cavitation only occurs in the apices and not in the bases. Yet the current conceptual model of tuberculosis renders the immunology of these two temporally and spatially separated events identical. One key consequence is that prevention of primary childhood tuberculosis at the lung bases is regarded as adequate immunological protection, but extensive evidence shows that greater immunity could predispose to immunopathology and transmission at the lung apex. A much greater understanding of time and place in the immunopathological mechanisms underlying human tuberculosis is needed before further pre-exposure vaccination trials can be done.
Collapse
Affiliation(s)
- Paul T Elkington
- Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, UK.
| | - Jon S Friedland
- Infectious Diseases and Immunity, Imperial College London, UK
| |
Collapse
|
152
|
Soares de Melo C, Candice SDM, Feng TS, van der Westhuyzen R, Gessner RK, Street LJ, Morgans GL, Warner DF, Moosa A, Naran K, Lawrence N, Boshoff HIM, Barry CE, Harris CJ, Gordon R, Chibale K. Aminopyrazolo[1,5-a]pyrimidines as potential inhibitors of Mycobacterium tuberculosis: Structure activity relationships and ADME characterization. Bioorg Med Chem 2015; 23:7240-50. [PMID: 26522089 DOI: 10.1016/j.bmc.2015.10.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/05/2015] [Accepted: 10/13/2015] [Indexed: 11/24/2022]
Abstract
Whole-cell high-throughput screening of a diverse SoftFocus library against Mycobacterium tuberculosis (Mtb) generated a novel aminopyrazolo[1,5-a]pyrimidine hit series. The synthesis and structure activity relationship studies identified compounds with potent antimycobacterial activity. The SAR of over 140 compounds shows that the 2-pyridylmethylamine moiety at the C-7 position of the pyrazolopyrimidine scaffold was important for Mtb activity, whereas the C-3 position offered a higher degree of flexibility. The series was also profiled for in vitro cytotoxicity and microsomal metabolic stability as well as physicochemical properties. Consequently liabilities to be addressed in a future lead optimization campaign have been identified.
Collapse
Affiliation(s)
| | - Soares de Melo Candice
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Tzu-Shean Feng
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Renier van der Westhuyzen
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Richard K Gessner
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Leslie J Street
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | | | - Digby F Warner
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Atica Moosa
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
| | - Krupa Naran
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
| | - Nina Lawrence
- Division of Clinical Pharmacology, University of Cape Town, Rondebosch 7701, South Africa
| | - Helena I M Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - C John Harris
- Cjh Consultants (Pharmaceutical R&D), Lydith, High Street, Eynsford, Kent DA4 0AB, United Kingdom
| | - Richard Gordon
- Strategic Health Innovation Partnerships (SHIP), South African Medical Research Council, Parow Valley, South Africa
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa; South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa.
| |
Collapse
|
153
|
Abstract
The world is in need of more effective approaches to controlling tuberculosis. The development of improved control strategies has been hampered by deficiencies in the tools available for detecting Mycobacterium tuberculosis and defining the dynamic consequences of the interaction of M. tuberculosis with its human host. Key needs include a highly sensitive, specific nonsputum diagnostic; biomarkers predictive of responses to therapy; correlates of risk for disease development; and host response-independent markers of M. tuberculosis infection. Tools able to sensitively detect and quantify total body M. tuberculosis burden might well be transformative across many needed use cases. Here, we review the current state of the field, paying particular attention to needed changes in experimental paradigms that would facilitate the discovery, validation, and development of such tools.
Collapse
Affiliation(s)
- Jennifer L Gardiner
- Discovery and Translational Sciences, Global Health, Bill & Melinda Gates Foundation, Seattle, WA 98102
| | - Christopher L Karp
- Discovery and Translational Sciences, Global Health, Bill & Melinda Gates Foundation, Seattle, WA 98102
| |
Collapse
|
154
|
E1 of α-ketoglutarate dehydrogenase defends Mycobacterium tuberculosis against glutamate anaplerosis and nitroxidative stress. Proc Natl Acad Sci U S A 2015; 112:E5834-43. [PMID: 26430237 DOI: 10.1073/pnas.1510932112] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Enzymes of central carbon metabolism (CCM) in Mycobacterium tuberculosis (Mtb) make an important contribution to the pathogen's virulence. Evidence is emerging that some of these enzymes are not simply playing the metabolic roles for which they are annotated, but can protect the pathogen via additional functions. Here, we found that deficiency of 2-hydroxy-3-oxoadipate synthase (HOAS), the E1 component of the α-ketoglutarate (α-KG) dehydrogenase complex (KDHC), did not lead to general metabolic perturbation or growth impairment of Mtb, but only to the specific inability to cope with glutamate anaplerosis and nitroxidative stress. In the former role, HOAS acts to prevent accumulation of aldehydes, including growth-inhibitory succinate semialdehyde (SSA). In the latter role, HOAS can participate in an alternative four-component peroxidase system, HOAS/dihydrolipoyl acetyl transferase (DlaT)/alkylhydroperoxide reductase colorless subunit gene (ahpC)-neighboring subunit (AhpD)/AhpC, using α-KG as a previously undescribed source of electrons for reductase action. Thus, instead of a canonical role in CCM, the E1 component of Mtb's KDHC serves key roles in situational defense that contribute to its requirement for virulence in the host. We also show that pyruvate decarboxylase (AceE), the E1 component of pyruvate dehydrogenase (PDHC), can participate in AceE/DlaT/AhpD/AhpC, using pyruvate as a source of electrons for reductase action. Identification of these systems leads us to suggest that Mtb can recruit components of its CCM for reactive nitrogen defense using central carbon metabolites.
Collapse
|
155
|
Garay CD, Dreyfuss JM, Galagan JE. Metabolic modeling predicts metabolite changes in Mycobacterium tuberculosis. BMC SYSTEMS BIOLOGY 2015; 9:57. [PMID: 26377923 PMCID: PMC4574064 DOI: 10.1186/s12918-015-0206-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 09/03/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Mycobacterium tuberculosis (MTB) is the causal agent of the disease tuberculosis (TB). Metabolic adaptations are thought to be critical to the survival of MTB during pathogenesis. Computational tools that can be used to study MTB metabolism in silico and prioritize resource-intensive experimental work could significantly accelerate research. RESULTS We have developed E-Flux-MFC, an enhancement of our original E-Flux method that enables the prediction of changes in the production of external and internal metabolites corresponding to gene expression measurements. We have used this method to simulate the changes in the metabolic state of Mycobacterium tuberculosis (MTB). We have validated the accuracy of E-Flux-MFC for predicting changes in lipids and metabolites during a hypoxia time course using previously published metabolomics and transcriptomics data. We have further validated the accuracy of the method for predicting changes in MTB lipids following the deletion and induction of two well-studied transcription factors (TFs). We have applied the method to predict the metabolic impact of the induction of each of the approximately 180 MTB TFs using a previously generated and publically available expression data set. CONCLUSIONS E-flux-MFC can be used to study global changes in MTB metabolites from gene expression data associated with environmental and genetic perturbations. The application of this method to a data set of MTB TF perturbations provides a resource for studying the large number of TFs whose functions remain unknown. Most TFs impact metabolites indirectly through the propagation of gene expression changes through the regulatory network rather than through their direct regulons. E-Flux-MFC is also applicable to any organism for which accurate metabolic models are available.
Collapse
Affiliation(s)
- Christopher D Garay
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA.
| | - Jonathan M Dreyfuss
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA. .,Joslin Diabetes Center, Boston, MA, 02215, USA.
| | - James E Galagan
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA. .,Graduate Program in Bioinformatics, Boston University, Boston, MA, 02215, USA. .,National Emerging Infectious Diseases Laboratories, Boston, MA, 02118, USA.
| |
Collapse
|
156
|
Huang J, Jiao J, Xu W, Zhao H, Zhang C, Shi Y, Xiao Z. MiR-155 is upregulated in patients with active tuberculosis and inhibits apoptosis of monocytes by targeting FOXO3. Mol Med Rep 2015; 12:7102-8. [PMID: 26324048 DOI: 10.3892/mmr.2015.4250] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 07/21/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the association between microRNA (miR)-155 and apoptosis of monocytes infected by Mycobacterium tuberculosis, to examine the effect of forkhead box O3 (FOXO3) on miR‑155. The present study analysed the apoptosis of CD14+ in the peripheral blood of patients with active tuberculosis, disposed the THP‑1 human monocytic cell line by BCG and examined the expression of miR‑155. Furthermore, the expression of FOXO3 in THP‑1 cells was determined, and wild- and mutant-type luciferase reporter plasmids containing FOXO3 3'‑untranslated regions (UTRs) were constructed to analyse the expression of luciferase. Finally, an over‑expression plasmid was constructed, and THP-1 cells were transfected with control miRNA, miR‑155 and the plasmid, which revealed that miR‑155 inhibited the apoptosis of THP‑1 cells. miR‑155 in the THP‑1 cells infected by BCG was upregulated and apoptosis also increased. However, the apoptosis declined when the cells were transfected with the control miRNA and miR‑155. Folllowing transfection with miR‑155, the expression of FOXO3 decreased. Transfection with miR‑155 and the FOXO3 3'-UTRs significantly reduced luciferase, and overexpression of FOXO3 reversed the inhibitory role of miR‑155. From these results, it was concluded that mycobacteria can improve the level of miR‑155, while BCG can induce apoptosis in THP‑1 cells. The results suggested FOXO3 is a downstream target gene of miR‑155, which combines 3'-UTRs to inhibit the expression of FOXO3.
Collapse
Affiliation(s)
- Jian Huang
- Second Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Junhua Jiao
- First Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Weihua Xu
- First Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Huayang Zhao
- Third Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Chunxiao Zhang
- Third Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Yan Shi
- Second Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Zhijian Xiao
- Second Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| |
Collapse
|
157
|
Datta M, Via LE, Chen W, Baish JW, Xu L, Barry CE, Jain RK. Mathematical Model of Oxygen Transport in Tuberculosis Granulomas. Ann Biomed Eng 2015; 44:863-72. [PMID: 26253038 DOI: 10.1007/s10439-015-1415-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/01/2015] [Indexed: 11/26/2022]
Abstract
Pulmonary granulomas--the hallmark of Mycobacterium tuberculosis (MTB) infection--are dense cellular lesions that often feature regions of hypoxia and necrosis, partially due to limited transport of oxygen. Low oxygen in granulomas can impair the host immune response, while MTB are able to adapt and persist in hypoxic environments. Here, we used a physiologically based mathematical model of oxygen diffusion and consumption to calculate oxygen profiles within the granuloma, assuming Michaelis-Menten kinetics. An approximate analytical solution--using a priori and newly estimated parameters from experimental data in a rabbit model of tuberculosis--was able to predict the size of hypoxic and necrotic regions in agreement with experimental results from the animal model. Such quantitative understanding of transport limitations can inform future tuberculosis therapeutic strategies that may include adjunct host-directed therapies that facilitate oxygen and drug delivery for more effective treatment.
Collapse
Affiliation(s)
- Meenal Datta
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Cox 7, 100 Blossom Street, Boston, MA, 02114, USA
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, 02155, USA
| | - Laura E Via
- Tuberculosis Research Section, Laboratory of Clinical Infectious Disease, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Clinical Laboratory Sciences, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Wei Chen
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Cox 7, 100 Blossom Street, Boston, MA, 02114, USA
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - James W Baish
- Departments of Biomedical and Mechanical Engineering, Bucknell University, Lewisburg, PA, 17837, USA
| | - Lei Xu
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Cox 7, 100 Blossom Street, Boston, MA, 02114, USA
| | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Infectious Disease, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Clinical Laboratory Sciences, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Cox 7, 100 Blossom Street, Boston, MA, 02114, USA.
| |
Collapse
|
158
|
Niu NK, Yin JJ, Yang YX, Wang ZL, Zhou ZW, He ZX, Chen XW, Zhang X, Duan W, Yang T, Zhou SF. Novel targeting of PEGylated liposomes for codelivery of TGF-β1 siRNA and four antitubercular drugs to human macrophages for the treatment of mycobacterial infection: a quantitative proteomic study. Drug Des Devel Ther 2015; 9:4441-70. [PMID: 26300629 PMCID: PMC4535548 DOI: 10.2147/dddt.s79369] [Citation(s) in RCA: 11] [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] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB) is still a major public health issue in developing countries, and its chemotherapy is compromised by poor drug compliance and severe side effects. This study aimed to synthesize and characterize new multimodal PEGylated liposomes encapsulated with clinically commonly used anti-TB drugs with linkage to small interfering RNA (siRNA) against transforming growth factor-β1 (TGF-β1). The novel NP-siRNA liposomes could target THP-1-derived human macrophages that were the host cells of mycobacterium infection. The biological effects of the NP-siRNA liposomes were evaluated on cell cycle distribution, apoptosis, autophagy, and the gene silencing efficiency of TGF-β1 siRNA in human macrophages. We also explored the proteomic responses to the newly synthesized NP-siRNA liposomes using the stable isotope labeling with amino acids in cell culture approach. The results showed that the multifunctional PEGylated liposomes were successfully synthesized and chemically characterized with a mean size of 265.1 nm. The novel NP-siRNA liposomes functionalized with the anti-TB drugs and TGF-β1 siRNA were endocytosed efficiently by human macrophages as visualized by transmission electron microscopy and scanning electron microscopy. Furthermore, the liposomes showed a low cytotoxicity toward human macrophages. There was no significant effect on cell cycle distribution and apoptosis in THP-1-derived macrophages after drug exposure at concentrations ranging from 2.5 to 62.5 μg/mL. Notably, there was a 6.4-fold increase in the autophagy of human macrophages when treated with the NP-siRNA liposomes at 62.5 μg/mL. In addition, the TGF-β1 and nuclear factor-κB expression levels were downregulated by the NP-siRNA liposomes in THP-1-derived macrophages. The Ingenuity Pathway Analysis data showed that there were over 40 signaling pathways involved in the proteomic responses to NP-siRNA liposome exposure in human macrophages, with 160 proteins mapped. The top five canonical signaling pathways were eukaryotic initiation factor 2 signaling, actin cytoskeleton signaling, remodeling of epithelial adherens junctions, epithelial adherens junction signaling, and Rho GDP-dissociation inhibitor signaling pathways. Collectively, the novel synthetic targeting liposomes represent a promising delivery system for anti-TB drugs to human macrophages with good selectivity and minimal cytotoxicity.
Collapse
Affiliation(s)
- Ning-Kui Niu
- Department of Orthopedics, General Hospital of Tianjin Medical University, Tianjin, People’s Republic of China
- Department of Spinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Juan-Juan Yin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Yin-Xue Yang
- Department of Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| | - Zi-Li Wang
- Department of Orthopedics, General Hospital of Tianjin Medical University, Tianjin, People’s Republic of China
| | - Zhi-Wei Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Zhi-Xu He
- Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Xiao-Wu Chen
- Department of General Surgery, The First People’s Hospital of Shunde Affiliated to Southern Medical University, Shunde, Foshan, Guangdong, People’s Republic of China
| | - Xueji Zhang
- Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, People’s Republic of China
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, VIC, Australia
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Salt Lake Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| |
Collapse
|
159
|
Mehra S, Foreman TW, Didier PJ, Ahsan MH, Hudock TA, Kissee R, Golden NA, Gautam US, Johnson AM, Alvarez X, Russell-Lodrigue KE, Doyle LA, Roy CJ, Niu T, Blanchard JL, Khader SA, Lackner AA, Sherman DR, Kaushal D. The DosR Regulon Modulates Adaptive Immunity and Is Essential for Mycobacterium tuberculosis Persistence. Am J Respir Crit Care Med 2015; 191:1185-96. [PMID: 25730547 DOI: 10.1164/rccm.201408-1502oc] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
RATIONALE Hypoxia promotes dormancy by causing physiologic changes to actively replicating Mycobacterium tuberculosis. DosR controls the response of M. tuberculosis to hypoxia. OBJECTIVES To understand DosR's contribution in the persistence of M. tuberculosis, we compared the phenotype of various DosR regulon mutants and a complemented strain to M. tuberculosis in macaques, which faithfully model M. tuberculosis infection. METHODS We measured clinical and microbiologic correlates of infection with M. tuberculosis relative to mutant/complemented strains in the DosR regulon, studied lung pathology and hypoxia, and compared immune responses in lung using transcriptomics and flow cytometry. MEASUREMENTS AND MAIN RESULTS Despite being able to replicate initially, mutants in DosR regulon failed to persist or cause disease. On the contrary, M. tuberculosis and a complemented strain were able to establish infection and tuberculosis. The attenuation of pathogenesis in animals infected with the mutants coincided with the appearance of a Th1 response and organization of hypoxic lesions wherein M. tuberculosis expressed dosR. The lungs of animals infected with the mutants (but not the complemented strain) exhibited early transcriptional signatures of T-cell recruitment, activation, and proliferation associated with an increase of T cells expressing homing and proliferation markers. CONCLUSIONS Delayed adaptive responses, a hallmark of M. tuberculosis infection, not only lead to persistence but also interfere with the development of effective antituberculosis vaccines. The DosR regulon therefore modulates both the magnitude and the timing of adaptive immune responses in response to hypoxia in vivo, resulting in persistent infection. Hence, DosR regulates key aspects of the M. tuberculosis life cycle and limits lung pathology.
Collapse
|
160
|
Sun J, Siroy A, Lokareddy RK, Speer A, Doornbos KS, Cingolani G, Niederweis M. The tuberculosis necrotizing toxin kills macrophages by hydrolyzing NAD. Nat Struct Mol Biol 2015; 22:672-8. [PMID: 26237511 PMCID: PMC4560639 DOI: 10.1038/nsmb.3064] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 07/06/2015] [Indexed: 12/29/2022]
Abstract
Mycobacterium tuberculosis (Mtb) induces necrosis of infected cells to evade immune responses. Recently, we found that Mtb uses the protein CpnT to kill human macrophages by secreting its C-terminal domain, named tuberculosis necrotizing toxin (TNT), which induces necrosis by an unknown mechanism. Here we show that TNT gains access to the cytosol of Mtb-infected macrophages, where it hydrolyzes the essential coenzyme NAD(+). Expression or injection of a noncatalytic TNT mutant showed no cytotoxicity in macrophages or in zebrafish zygotes, respectively, thus demonstrating that the NAD(+) glycohydrolase activity is required for TNT-induced cell death. To prevent self-poisoning, Mtb produces an immunity factor for TNT (IFT) that binds TNT and inhibits its activity. The crystal structure of the TNT-IFT complex revealed a new NAD(+) glycohydrolase fold of TNT, the founding member of a toxin family widespread in pathogenic microorganisms.
Collapse
Affiliation(s)
- Jim Sun
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Axel Siroy
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ravi K Lokareddy
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Alexander Speer
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kathryn S Doornbos
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Gino Cingolani
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Michael Niederweis
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
161
|
Fronczek CF, Yoon JY. Biosensors for Monitoring Airborne Pathogens. ACTA ACUST UNITED AC 2015; 20:390-410. [DOI: 10.1177/2211068215580935] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Indexed: 01/15/2023]
|
162
|
Abstract
SUMMARY Tuberculosis (TB) is a leading cause of death worldwide despite the availability of effective chemotherapy for over 60 years. Although Mycobacterium bovis bacillus Calmette-Guérin (BCG) vaccination protects against active TB disease in some populations, its efficacy is suboptimal. Development of an effective TB vaccine is a top global priority that has been hampered by an incomplete understanding of protective immunity to TB. Thus far, preventing TB disease, rather than infection, has been the primary target for vaccine development. Several areas of research highlight the importance of including preinfection vaccines in the development pipeline. First, epidemiology and mathematical modeling studies indicate that a preinfection vaccine would have a high population-level impact for control of TB disease. Second, immunology studies support the rationale for targeting prevention of infection, with evidence that host responses may be more effective during acute infection than during chronic infection. Third, natural history studies indicate that resistance to TB infection occurs in a small percentage of the population. Fourth, case-control studies of BCG indicate that it may provide protection from infection. Fifth, prevention-of-infection trials would have smaller sample sizes and a shorter duration than disease prevention trials and would enable opportunities to search for correlates of immunity as well as serve as a criterion for selecting a vaccine product for testing in a larger TB disease prevention trial. Together, these points support expanding the focus of TB vaccine development efforts to include prevention of infection as a primary goal along with vaccines or other interventions that reduce the rate of transmission and reactivation.
Collapse
|
163
|
Rustad TR, Minch KJ, Ma S, Winkler JK, Hobbs S, Hickey M, Brabant W, Turkarslan S, Price ND, Baliga NS, Sherman DR. Mapping and manipulating the Mycobacterium tuberculosis transcriptome using a transcription factor overexpression-derived regulatory network. Genome Biol 2015; 15:502. [PMID: 25380655 DOI: 10.1186/preaccept-1701638048134699] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mycobacterium tuberculosis senses and responds to the shifting and hostile landscape of the host. To characterize the underlying intertwined gene regulatory network governed by approximately 200 transcription factors of M. tuberculosis, we have assayed the global transcriptional consequences of overexpressing each transcription factor from an inducible promoter. RESULTS We cloned and overexpressed 206 transcription factors in M. tuberculosis to identify the regulatory signature of each. We identified 9,335 regulatory consequences of overexpressing each of 183 transcription factors, providing evidence of regulation for 70% of the M. tuberculosis genome. These transcriptional signatures agree well with previously described M. tuberculosis regulons. The number of genes differentially regulated by transcription factor overexpression varied from hundreds of genes to none, with the majority of expression changes repressing basal transcription. Exploring the global transcriptional maps of transcription factor overexpressing (TFOE) strains, we predicted and validated the phenotype of a regulator that reduces susceptibility to a first line anti-tubercular drug, isoniazid. We also combined the TFOE data with an existing model of M. tuberculosis metabolism to predict the growth rates of individual TFOE strains with high fidelity. CONCLUSION This work has led to a systems-level framework describing the transcriptome of a devastating bacterial pathogen, characterized the transcriptional influence of nearly all individual transcription factors in M. tuberculosis, and demonstrated the utility of this resource. These results will stimulate additional systems-level and hypothesis-driven efforts to understand M. tuberculosis adaptations that promote disease.
Collapse
|
164
|
Rustad TR, Minch KJ, Ma S, Winkler JK, Hobbs S, Hickey M, Brabant W, Turkarslan S, Price ND, Baliga NS, Sherman DR. Mapping and manipulating the Mycobacterium tuberculosis transcriptome using a transcription factor overexpression-derived regulatory network. Genome Biol 2015. [PMID: 25380655 PMCID: PMC4249609 DOI: 10.1186/s13059-014-0502-3] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Mycobacterium tuberculosis senses and responds to the shifting and hostile landscape of the host. To characterize the underlying intertwined gene regulatory network governed by approximately 200 transcription factors of M. tuberculosis, we have assayed the global transcriptional consequences of overexpressing each transcription factor from an inducible promoter. Results We cloned and overexpressed 206 transcription factors in M. tuberculosis to identify the regulatory signature of each. We identified 9,335 regulatory consequences of overexpressing each of 183 transcription factors, providing evidence of regulation for 70% of the M. tuberculosis genome. These transcriptional signatures agree well with previously described M. tuberculosis regulons. The number of genes differentially regulated by transcription factor overexpression varied from hundreds of genes to none, with the majority of expression changes repressing basal transcription. Exploring the global transcriptional maps of transcription factor overexpressing (TFOE) strains, we predicted and validated the phenotype of a regulator that reduces susceptibility to a first line anti-tubercular drug, isoniazid. We also combined the TFOE data with an existing model of M. tuberculosis metabolism to predict the growth rates of individual TFOE strains with high fidelity. Conclusion This work has led to a systems-level framework describing the transcriptome of a devastating bacterial pathogen, characterized the transcriptional influence of nearly all individual transcription factors in M. tuberculosis, and demonstrated the utility of this resource. These results will stimulate additional systems-level and hypothesis-driven efforts to understand M. tuberculosis adaptations that promote disease. Electronic supplementary material The online version of this article (doi:10.1186/s13059-014-0502-3) contains supplementary material, which is available to authorized users.
Collapse
|
165
|
Rayasam GV, Balganesh TS. Exploring the potential of adjunct therapy in tuberculosis. Trends Pharmacol Sci 2015; 36:506-13. [PMID: 26073420 DOI: 10.1016/j.tips.2015.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/15/2015] [Accepted: 05/15/2015] [Indexed: 01/02/2023]
Abstract
A critical unmet need for treatment of drug-resistant tuberculosis (TB) is to find novel therapies that are efficacious, safe, and shorten the duration of treatment. Drug discovery approaches for TB primarily target essential genes of the pathogen Mycobacterium tuberculosis (Mtb) but novel strategies such as host-directed therapies and nonmicrobicidal targets are necessary to bring about a paradigm shift in treatment. Drugs targeting the host pathways and nonmicrobicidal proteins can be used only in conjunction with existing drugs as adjunct therapies. Significantly, host-directed adjunct therapies have the potential to decrease duration of treatment, as they are less prone to drug resistance, target the immune responses, and act via novel mechanism of action. Recent advances in targeting host-pathogen interactions have implicated pathways such as eicosanoid regulation and angiogenesis. Furthermore, several approved drugs such as metformin and verapamil have been identified that appear suitable for repurposing for the treatment of TB. These findings and the challenges in the area of host- and/or pathogen-directed adjunct therapies and their implications for TB therapy are discussed.
Collapse
Affiliation(s)
- Geetha Vani Rayasam
- CSIR-Open Source Drug Discovery (OSDD) Unit, Council for Scientific and Industrial Research, Anusandhan Bhavan, 2 Rafi Marg, New Delhi, India.
| | | |
Collapse
|
166
|
Emmadi NR, Bingi C, Kotapalli SS, Ummanni R, Nanubolu JB, Atmakur K. Synthesis and evaluation of novel fluorinated pyrazolo-1,2,3-triazole hybrids as antimycobacterial agents. Bioorg Med Chem Lett 2015; 25:2918-22. [PMID: 26048808 DOI: 10.1016/j.bmcl.2015.05.044] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/18/2015] [Accepted: 05/20/2015] [Indexed: 12/20/2022]
Abstract
A library of novel 3-trifluoromethyl pyrazolo-1,2,3-triazole hybrids (5-7) were accomplished starting from 5-phenyl-3-(trifluoromethyl)-1H-pyrazol-4-amine (1) via key intermediate 2-azido-N-(5-phenyl-3-(trifluoromethyl)-1H-pyrazol-4-yl)acetamide (3) through click chemistry approach. Thus obtained compounds in 5-7 series were evaluated for in vitro antimycobacterial activity against Mycobacterium smegmatis (MC(2) 155) and also verified the cytotoxicity. These studies engendered promising lead compounds 5q, 7b and 7c with MIC (μg/mL) values 15.34, 16.18 and 16.60, respectively. Amongst these three compounds, 2-(4-(4-methoxybenzoyl)-1H-1,2,3-triazol-1-yl)-N-(5-phenyl-3-(trifluoromethyl)-1H-pyrazol-4-yl) acetamide (5q) emerged as the most promising antitubercular agent with lowest cytotoxicity against the A549 cancer cell line. This is the first report to demonstrate the pyrazolo triazole hybrids as potential antimycobacterial agents.
Collapse
Affiliation(s)
- Narender Reddy Emmadi
- Crop Protection Chemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Chiranjeevi Bingi
- Crop Protection Chemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Sudha Sravanti Kotapalli
- Chemical Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Ramesh Ummanni
- Chemical Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Jagadeesh Babu Nanubolu
- Centre for X-ray Crystallography, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Krishnaiah Atmakur
- Crop Protection Chemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India.
| |
Collapse
|
167
|
Lee WH, Loo CY, Traini D, Young PM. Nano- and micro-based inhaled drug delivery systems for targeting alveolar macrophages. Expert Opin Drug Deliv 2015; 12:1009-26. [PMID: 25912721 DOI: 10.1517/17425247.2015.1039509] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Macrophages are the most versatile cells in the hematopoietic system and are strategically distributed in tissues to fight pathogens or other foreign particles. In the lung, however, for intracellular infections such as tuberculosis, pneumonia and aspergillosis, bacteria and fungi utilize the alveolar macrophage as a breeding ground. This has become a challenge for the treatment of these infections, as most drugs do not effectively reach the macrophages at therapeutic levels. Alveolar macrophages also play an important role to initiative adaptive immunity toward combating inflammation and cancer in the lung. AREAS COVERED This review focuses on the development of micro- and nanotechnology-based drug delivery systems to target alveolar macrophages in association with intracellular infections, cancer and lung inflammation. Aspects of nanoparticle and micron-sized particle engineering through exploitation of particles' physicochemical characteristics such as particle size, surface charge and geometry of particles are discussed. In addition, the application of nanocarriers such as liposomes, polymeric nanoparticles and dendrimers are covered with respect to macrophage targeting. EXPERT OPINION Drug delivery targeted to alveolar macrophages in the lung is becoming a reality thanks to micro- and nanotechnology breakthrough. The literature review shows that regulation of physicochemical parameters of particles could be a recipe to enhance macrophage targeting and uptake. However, there is still a need to identify more target-specific receptors in order to facilitate drug targeting. Besides that, the toxicity of nanocarriers arising from prolonged residence in the lung should be taken into consideration during formulation.
Collapse
Affiliation(s)
- Wing-Hin Lee
- Woolcock Institute of Medical Research, Sydney Medical School, Respiratory Technology, The Discipline of Pharmacology , Sydney, 2006 , Australia
| | | | | | | |
Collapse
|
168
|
Elliott TOJP, Owolabi O, Donkor S, Kampmann B, Hill PC, Ottenhoff THM, Haks MC, Kaufmann SHE, Maertzdorf J, Sutherland JS. Dysregulation of Apoptosis Is a Risk Factor for Tuberculosis Disease Progression. J Infect Dis 2015; 212:1469-79. [PMID: 25895988 DOI: 10.1093/infdis/jiv238] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/25/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND A major barrier to effective tuberculosis control is our limited understanding of risk factors for tuberculosis disease progression. This study examined the role of apoptosis in immunity to tuberculosis. METHODS Cell subsets from tuberculosis cases and tuberculin skin test-positive (TST(+)) and TST-negative (TST(-)) household contacts (HHCs) were analyzed for expression of annexin-V and propidium iodide by flow cytometry. RNA microarrays were used to determine differences in apoptotic gene expression levels and multiplex ligation-dependent probe amplification was used to analyze gene expression in HHCs who progressed to active tuberculosis. RESULTS T cells from TST(+)HHC exhibited higher levels of apoptosis than tuberculosis cases; however, tuberculosis cases had a higher proportion of late apoptotic cells within the CD3(+)PD-1(+) subset. Tuberculosis cases had reduced levels of antiapoptotic genes compared to HHCs with a significant reduction in BCL2 associated with disease progression at least 1 year prior to progression. CONCLUSIONS While T cells are clearly able to mount a robust immune response to Mycobacterium tuberculosis, there are increased levels of apoptosis seen in effector T cells from tuberculosis patients. Dysregulation of several apoptotic genes suggest that apoptosis is a major functional pathway that could be targeted for future host-directed therapeutics.
Collapse
Affiliation(s)
- Thomas O J P Elliott
- Vaccines & Immunity, Medical Research Council Unit, Fajara, The Gambia University of Manchester, United Kingdom
| | - Olumuyiwa Owolabi
- Vaccines & Immunity, Medical Research Council Unit, Fajara, The Gambia
| | - Simon Donkor
- Vaccines & Immunity, Medical Research Council Unit, Fajara, The Gambia
| | - Beate Kampmann
- Vaccines & Immunity, Medical Research Council Unit, Fajara, The Gambia
| | - Philip C Hill
- Vaccines & Immunity, Medical Research Council Unit, Fajara, The Gambia Centre for International Health, Department of Preventive and Social Medicine, Faculty of Medicine, University of Otago, Dunedin, New Zealand
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, The Netherlands
| | - Marielle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, The Netherlands
| | - Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute of Infection Biology, Berlin, Germany
| | - Jeroen Maertzdorf
- Department of Immunology, Max Planck Institute of Infection Biology, Berlin, Germany
| | | |
Collapse
|
169
|
Chakraborty S, Rhee KY. Tuberculosis Drug Development: History and Evolution of the Mechanism-Based Paradigm. Cold Spring Harb Perspect Med 2015; 5:a021147. [PMID: 25877396 DOI: 10.1101/cshperspect.a021147] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Modern tuberculosis (TB) chemotherapy is widely viewed as a crowning triumph of anti-infectives research. However, only one new TB drug has entered clinical practice in the past 40 years while drug resistance threatens to further destabilize the pandemic. Here, we review a brief history of TB drug development, focusing on the evolution of mechanism(s)-of-action studies and key conceptual barriers to rational, mechanism-based drugs.
Collapse
Affiliation(s)
- Sumit Chakraborty
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medical College, New York, New York 10021 Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York 10021
| | - Kyu Y Rhee
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medical College, New York, New York 10021 Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York 10021
| |
Collapse
|
170
|
Shukla H, Kumar V, Singh AK, Rastogi S, Khan SR, Siddiqi MI, Krishnan MY, Akhtar MS. Isocitrate lyase of Mycobacterium tuberculosis is inhibited by quercetin through binding at N-terminus. Int J Biol Macromol 2015; 78:137-41. [PMID: 25869309 DOI: 10.1016/j.ijbiomac.2015.04.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 04/03/2015] [Accepted: 04/04/2015] [Indexed: 12/01/2022]
Abstract
Combating tuberculosis requires new therapeutic strategies that not only target the actively dividing bacilli but also the dormant bacilli during persistent infection. Isocitrate lyase (ICL) is a key enzyme of the glyoxylate shunt, crucial for the survival of bacteria in macrophages and mice. MtbICL is considered as one of the potential and attractive drug targets against persistent infection. We report the inhibition of MtbICL by quercetin with IC50 of 3.57 μM. In addition, quercetin strongly inhibited the growth of Mtb H37Rv utilizing acetate, rather than glucose as the sole carbon source, suggesting the inhibition of glyoxylate shunt. Quercetin binds at the N-terminus of MtbICL (Kd - 6.68 μM).
Collapse
Affiliation(s)
- Harish Shukla
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow 226 031, India
| | - Vikash Kumar
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow 226 031, India
| | - Amit Kumar Singh
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow 226 031, India
| | - Shivangi Rastogi
- Microbiology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow 226 031, India
| | - Shaheb Raj Khan
- Microbiology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow 226 031, India
| | - Mohammad Imran Siddiqi
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow 226 031, India; Academy of Scientific and Innovative Research, Chennai 600 113, India
| | - Manju Yasoda Krishnan
- Microbiology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow 226 031, India; Academy of Scientific and Innovative Research, Chennai 600 113, India
| | - Md Sohail Akhtar
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow 226 031, India; Academy of Scientific and Innovative Research, Chennai 600 113, India.
| |
Collapse
|
171
|
't Hart BA, Bogers WM, Haanstra KG, Verreck FA, Kocken CH. The translational value of non-human primates in preclinical research on infection and immunopathology. Eur J Pharmacol 2015; 759:69-83. [PMID: 25814254 DOI: 10.1016/j.ejphar.2015.03.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/09/2015] [Accepted: 03/12/2015] [Indexed: 01/01/2023]
Abstract
The immune system plays a central role in the defense against environmental threats - such as infection with viruses, parasites or bacteria - but can also be a cause of disease, such as in the case of allergic or autoimmune disorders. In the past decades the impressive development of biotechnology has provided scientists with biological tools for the development of highly selective treatments for the different types of disorders. However, despite some clear successes the translation of scientific discoveries into effective treatments has remained challenging. The often-disappointing predictive validity of the preclinical animal models that are used in the selection of the most promising vaccine or drug candidates is the Achilles heel in the therapy development process. This publication summarizes the relevance and usage of non-human primates as pre-clinical model in infectious and autoimmune diseases, in particular for biologicals, which due to their high species-specificity are inactive in lower species.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands; University of Groningen, University Medical Center, Department Neuroscience, Groningen, The Netherlands.
| | - Willy M Bogers
- Department Virology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | - Krista G Haanstra
- Department Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | - Frank A Verreck
- Department Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | - Clemens H Kocken
- Department Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| |
Collapse
|
172
|
Salunke SB, Azad AK, Kapuriya NP, Balada-Llasat JM, Pancholi P, Schlesinger LS, Chen CS. Design and synthesis of novel anti-tuberculosis agents from the celecoxib pharmacophore. Bioorg Med Chem 2015; 23:1935-43. [PMID: 25818768 DOI: 10.1016/j.bmc.2015.03.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/03/2015] [Accepted: 03/15/2015] [Indexed: 11/15/2022]
Abstract
The identification of compounds with anti-mycobacterial activity within classes of molecules that have been developed for other purposes is a fruitful approach for the development of anti-tuberculosis (TB) agents. In this study we used the scaffold of celecoxib which exhibits several activities against different pathogens, for the design and focused synthesis of a library of 64 compounds. For the primary screen, we used a bioluminescence-based method by constructing a luciferase-expressing reporter M.tb strain which contains the entire bacterial Lux operon cloned in a mycobacterial integrative expression vector. Through the screening of this library, we identified 6 hit compounds with high in vitro anti-mycobacterial activity (IC₅₀ ∼0.18-0.48 μM). In particular, compounds 41, 51 and 53 were capable of inhibiting M.tb as effectively as the anti-TB drug isoniazid (INH) at 5 μM over a 72-h period, as analyzed by both bioluminescence- and colony forming unit (CFU)-based assays. All hit compounds also showed anti-M.tb activities against several multi-drug-resistant (MDR) strains. Most of the hit compounds showed no cytotoxicity for human macrophages at concentrations as high as 40 μM, setting the stage for further optimization and development of these anti-TB hit compounds both ex vivo and in vivo.
Collapse
Affiliation(s)
- Santosh B Salunke
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, United States
| | - Abul K Azad
- Center for Microbial Interface Biology, The Ohio State University, 460 W 12th Avenue, Columbus, OH 43210, United States; Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Wexner Medical Center, 460 W 12th Avenue, Columbus, OH 43210, United States
| | - Naval P Kapuriya
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, United States; Division of Pharmaceutical and Organic Chemistry, M. & N. Virani Science College, Saurashtra University, Rajkot 360005, Gujarat, India
| | - Joan-Miquel Balada-Llasat
- Department of Pathology, The Ohio State University, 410 W. 10th Avenue, Columbus, OH 43210, United States
| | - Preeti Pancholi
- Department of Pathology, The Ohio State University, 410 W. 10th Avenue, Columbus, OH 43210, United States
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, The Ohio State University, 460 W 12th Avenue, Columbus, OH 43210, United States; Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Wexner Medical Center, 460 W 12th Avenue, Columbus, OH 43210, United States.
| | - Ching-Shih Chen
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, United States; Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
173
|
Nair V, Okello MO, Mangu NK, Seo BI, Gund MG. A novel molecule with notable activity against multi-drug resistant tuberculosis. Bioorg Med Chem Lett 2015; 25:1269-73. [PMID: 25677656 PMCID: PMC4348211 DOI: 10.1016/j.bmcl.2015.01.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 11/15/2022]
Abstract
Multi-drug resistant tuberculosis (MDR-TB) is emerging as a serious global health problem, which has been elevated through co-infection involving HIV and MDR-Mtb. The discovery of new compounds with anti-MDR TB efficacy and favorable metabolism profiles is an important scientific challenge. Using computational biology and ligand docking data, we have conceived a multifunctional molecule, 2, as a potential anti-MDR TB agent. This compound was produced through a multi-step synthesis. It exhibited significant in vitro activity against MDR-TB (MIC 1.56μg/mL) and its half-life (t1/2) in human liver microsomes was 14.4h. The metabolic profiles of compound 2 with respect to human cytochrome P450 (CYP) and uridine 5'-diphospho-glucuronosyltransferase (UGT) isozymes were favorable. Compound 2 also had relatively low in vitro cytotoxicity in uninfected macrophages. It displayed synergistic behavior against MDR-TB in combination with PA-824. Interestingly, compound 2 also displayed in vitro anti-HIV activity.
Collapse
Affiliation(s)
- Vasu Nair
- UGA Center for Drug Discovery and the College of Pharmacy, University of Georgia, R.C. Wilson Pharmacy, Room 320A, Athens, GA 30602, USA.
| | - Maurice O Okello
- UGA Center for Drug Discovery and the College of Pharmacy, University of Georgia, R.C. Wilson Pharmacy, Room 320A, Athens, GA 30602, USA
| | - Naveen K Mangu
- UGA Center for Drug Discovery and the College of Pharmacy, University of Georgia, R.C. Wilson Pharmacy, Room 320A, Athens, GA 30602, USA
| | - Byung I Seo
- UGA Center for Drug Discovery and the College of Pharmacy, University of Georgia, R.C. Wilson Pharmacy, Room 320A, Athens, GA 30602, USA
| | - Machhindra G Gund
- UGA Center for Drug Discovery and the College of Pharmacy, University of Georgia, R.C. Wilson Pharmacy, Room 320A, Athens, GA 30602, USA
| |
Collapse
|
174
|
Markad SD, Kaur P, Kishore Reddy BK, Chinnapattu M, Raichurkar A, Nandishaiah R, Panda M, Iyer PS. Novel lead generation of an anti-tuberculosis agent active against non-replicating mycobacteria: exploring hybridization of pyrazinamide with multiple fragments. Med Chem Res 2015. [DOI: 10.1007/s00044-015-1352-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
175
|
Gastelum-Aviña P, Velazquez C, Espitia C, Lares-Villa F, Garibay-Escobar A. A PE_PGRS33 protein ofMycobacterium tuberculosis: an ideal target for future tuberculosis vaccine design. Expert Rev Vaccines 2015; 14:699-711. [DOI: 10.1586/14760584.2015.1015995] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
176
|
Al Shammari B, Shiomi T, Tezera L, Bielecka MK, Workman V, Sathyamoorthy T, Mauri F, Jayasinghe SN, Robertson BD, D'Armiento J, Friedland JS, Elkington PT. The Extracellular Matrix Regulates Granuloma Necrosis in Tuberculosis. J Infect Dis 2015; 212:463-73. [PMID: 25676469 DOI: 10.1093/infdis/jiv076] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 01/29/2015] [Indexed: 12/30/2022] Open
Abstract
A central tenet of tuberculosis pathogenesis is that caseous necrosis leads to extracellular matrix destruction and bacterial transmission. We reconsider the underlying mechanism of tuberculosis pathology and demonstrate that collagen destruction may be a critical initial event, causing caseous necrosis as opposed to resulting from it. In human tuberculosis granulomas, regions of extracellular matrix destruction map to areas of caseous necrosis. In mice, transgenic expression of human matrix metalloproteinase 1 causes caseous necrosis, the pathological hallmark of human tuberculosis. Collagen destruction is the principal pathological difference between humanised mice and wild-type mice with tuberculosis, whereas the release of proinflammatory cytokines does not differ, demonstrating that collagen breakdown may lead to cell death and caseation. To investigate this hypothesis, we developed a 3-dimensional cell culture model of tuberculosis granuloma formation, using bioelectrospray technology. Collagen improved survival of Mycobacterium tuberculosis-infected cells analyzed on the basis of a lactate dehydrogenase release assay, propidium iodide staining, and measurement of the total number of viable cells. Taken together, these findings suggest that collagen destruction is an initial event in tuberculosis immunopathology, leading to caseous necrosis and compromising the immune response, revealing a previously unappreciated role for the extracellular matrix in regulating the host-pathogen interaction.
Collapse
Affiliation(s)
- Basim Al Shammari
- Infectious Diseases and Immunity Section, Division of Infectious Diseases
| | - Takayuki Shiomi
- Department of Medicine, Columbia University, New York, New York
| | - Liku Tezera
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine
| | - Magdalena K Bielecka
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine
| | - Victoria Workman
- BioPhysics Group, Department of Mechanical Engineering Institute of Biomedical Engineering Centre for Stem Cells and Regenerative Medicine, University College London
| | | | - Francesco Mauri
- Histopathology Department, Centre for Pathology, Division of Experimental Medicine
| | - Suwan N Jayasinghe
- BioPhysics Group, Department of Mechanical Engineering Institute of Biomedical Engineering Centre for Stem Cells and Regenerative Medicine, University College London
| | - Brian D Robertson
- MRC Centre for Molecular Bacteriology and Infection, Department of Medicine, Imperial College London
| | | | - Jon S Friedland
- Infectious Diseases and Immunity Section, Division of Infectious Diseases
| | - Paul T Elkington
- Infectious Diseases and Immunity Section, Division of Infectious Diseases NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine Institute for Life Sciences, University of Southampton, United Kingdom
| |
Collapse
|
177
|
Novel inhibitors of cholesterol degradation in Mycobacterium tuberculosis reveal how the bacterium's metabolism is constrained by the intracellular environment. PLoS Pathog 2015; 11:e1004679. [PMID: 25675247 PMCID: PMC4335503 DOI: 10.1371/journal.ppat.1004679] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/12/2015] [Indexed: 12/02/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) relies on a specialized set of metabolic pathways to support growth in macrophages. By conducting an extensive, unbiased chemical screen to identify small molecules that inhibit Mtb metabolism within macrophages, we identified a significant number of novel compounds that limit Mtb growth in macrophages and in medium containing cholesterol as the principle carbon source. Based on this observation, we developed a chemical-rescue strategy to identify compounds that target metabolic enzymes involved in cholesterol metabolism. This approach identified two compounds that inhibit the HsaAB enzyme complex, which is required for complete degradation of the cholesterol A/B rings. The strategy also identified an inhibitor of PrpC, the 2-methylcitrate synthase, which is required for assimilation of cholesterol-derived propionyl-CoA into the TCA cycle. These chemical probes represent new classes of inhibitors with novel modes of action, and target metabolic pathways required to support growth of Mtb in its host cell. The screen also revealed a structurally-diverse set of compounds that target additional stage(s) of cholesterol utilization. Mutants resistant to this class of compounds are defective in the bacterial adenylate cyclase Rv1625/Cya. These data implicate cyclic-AMP (cAMP) in regulating cholesterol utilization in Mtb, and are consistent with published reports indicating that propionate metabolism is regulated by cAMP levels. Intriguingly, reversal of the cholesterol-dependent growth inhibition caused by this subset of compounds could be achieved by supplementing the media with acetate, but not with glucose, indicating that Mtb is subject to a unique form of metabolic constraint induced by the presence of cholesterol. Human beings are the sole ecological niche for M. tuberculosis (Mtb), and it is estimated that 1.8 billion people are currently infected with Mtb. An important aspect of this infection is Mtb’s ability to maintain infection by replicating within macrophages. Within macrophages, Mtb exploits a specialized set of metabolic pathways to utilize host-derived nutrients, such as fatty acids and/or cholesterol, for energy production. Many details regarding Mtb metabolism during infection remain unknown. Here we took a chemical approach to identify small molecule probes, which target Mtb metabolism during infection in macrophages. We found that many of the small molecule inhibitors that we identified require cholesterol for activity. Here we report a novel chemical rescue approach to identify the metabolic targets of three novel inhibitors, and discovered that cAMP signaling is linked to cholesterol utilization in Mtb. Together, these data demonstrate that cholesterol exerts a dominant effect on Mtb metabolism within macrophages. Additionally, the novel inhibitors identified in this study will facilitate evaluation of cholesterol metabolism as a target for chemotherapeutic intervention.
Collapse
|
178
|
Ordas A, Raterink RJ, Cunningham F, Jansen HJ, Wiweger MI, Jong-Raadsen S, Bos S, Bates RH, Barros D, Meijer AH, Vreeken RJ, Ballell-Pages L, Dirks RP, Hankemeier T, Spaink HP. Testing tuberculosis drug efficacy in a zebrafish high-throughput translational medicine screen. Antimicrob Agents Chemother 2015; 59:753-62. [PMID: 25385118 PMCID: PMC4335901 DOI: 10.1128/aac.03588-14] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 11/05/2014] [Indexed: 12/17/2022] Open
Abstract
The translational value of zebrafish high-throughput screens can be improved when more knowledge is available on uptake characteristics of potential drugs. We investigated reference antibiotics and 15 preclinical compounds in a translational zebrafish-rodent screening system for tuberculosis. As a major advance, we have developed a new tool for testing drug uptake in the zebrafish model. This is important, because despite the many applications of assessing drug efficacy in zebrafish research, the current methods for measuring uptake using mass spectrometry do not take into account the possible adherence of drugs to the larval surface. Our approach combines nanoliter sampling from the yolk using a microneedle, followed by mass spectrometric analysis. To date, no single physicochemical property has been identified to accurately predict compound uptake; our method offers a great possibility to monitor how any novel compound behaves within the system. We have correlated the uptake data with high-throughput drug-screening data from Mycobacterium marinum-infected zebrafish larvae. As a result, we present an improved zebrafish larva drug-screening platform which offers new insights into drug efficacy and identifies potential false negatives and drugs that are effective in zebrafish and rodents. We demonstrate that this improved zebrafish drug-screening platform can complement conventional models of in vivo Mycobacterium tuberculosis-infected rodent assays. The detailed comparison of two vertebrate systems, fish and rodent, may give more predictive value for efficacy of drugs in humans.
Collapse
Affiliation(s)
- Anita Ordas
- IBL, Leiden University, Leiden, The Netherlands
| | - Robert-Jan Raterink
- Division of Analytical BioSciences, Leiden Academic Centre for Drug Research and Netherlands Metabolomics Centre, Leiden University, Leiden, The Netherlands
| | | | | | - Malgorzata I Wiweger
- IBL, Leiden University, Leiden, The Netherlands ZF-screens B.V., Leiden, The Netherlands
| | | | - Sabine Bos
- Division of Analytical BioSciences, Leiden Academic Centre for Drug Research and Netherlands Metabolomics Centre, Leiden University, Leiden, The Netherlands
| | | | | | | | - Rob J Vreeken
- Division of Analytical BioSciences, Leiden Academic Centre for Drug Research and Netherlands Metabolomics Centre, Leiden University, Leiden, The Netherlands
| | | | | | - Thomas Hankemeier
- Division of Analytical BioSciences, Leiden Academic Centre for Drug Research and Netherlands Metabolomics Centre, Leiden University, Leiden, The Netherlands
| | | |
Collapse
|
179
|
Anti-vascular endothelial growth factor treatment normalizes tuberculosis granuloma vasculature and improves small molecule delivery. Proc Natl Acad Sci U S A 2015; 112:1827-32. [PMID: 25624495 DOI: 10.1073/pnas.1424563112] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tuberculosis (TB) causes almost 2 million deaths annually, and an increasing number of patients are resistant to existing therapies. Patients who have TB require lengthy chemotherapy, possibly because of poor penetration of antibiotics into granulomas where the bacilli reside. Granulomas are morphologically similar to solid cancerous tumors in that they contain hypoxic microenvironments and can be highly fibrotic. Here, we show that TB-infected rabbits have impaired small molecule distribution into these disease sites due to a functionally abnormal vasculature, with a low-molecular-weight tracer accumulating only in peripheral regions of granulomatous lesions. Granuloma-associated vessels are morphologically and spatially heterogeneous, with poor vessel pericyte coverage in both human and experimental rabbit TB granulomas. Moreover, we found enhanced VEGF expression in both species. In tumors, antiangiogenic, specifically anti-VEGF, treatments can "normalize" their vasculature, reducing hypoxia and creating a window of opportunity for concurrent chemotherapy; thus, we investigated vessel normalization in rabbit TB granulomas. Treatment of TB-infected rabbits with the anti-VEGF antibody bevacizumab significantly decreased the total number of vessels while normalizing those vessels that remained. As a result, hypoxic fractions of these granulomas were reduced and small molecule tracer delivery was increased. These findings demonstrate that bevacizumab treatment promotes vascular normalization, improves small molecule delivery, and decreases hypoxia in TB granulomas, thereby providing a potential avenue to improve delivery and efficacy of current treatment regimens.
Collapse
|
180
|
Yihao D, Hongyun H, Maodan T. Latency-associated protein Rv2660c of Mycobacterium tuberculosis augments expression of proinflammatory cytokines in human macrophages by interacting with TLR2. Infect Dis (Lond) 2015; 47:168-77. [DOI: 10.3109/00365548.2014.982167] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
181
|
Shukla H, Kumar V, Singh AK, Singh N, Kashif M, Siddiqi MI, Yasoda Krishnan M, Sohail Akhtar M. Insight into the structural flexibility and function of Mycobacterium tuberculosis isocitrate lyase. Biochimie 2015; 110:73-80. [PMID: 25576828 DOI: 10.1016/j.biochi.2014.12.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/26/2014] [Indexed: 10/24/2022]
Abstract
Isocitrate lyase (ICL), is a key enzyme of the glyoxylate shunt crucial for the survival of Mycobacterium tuberculosis (Mtb) in macrophages during persistent infection. MtbICL catalyses the first step of this carbon anaplerosis cycle and is considered as a potential anti-tubercular drug target. The MtbICL is a tetramer with 222 symmetry, and each subunit of the enzymeis composed of 14 α-helices and 14 β-strands. We studied the conformational flexibility of the enzyme to get a deeper insight into its stability and function. Our studies show that the mutation of His180, close to the MtbICL signature sequence (K193KCGH197) completely abolishes the oligomeric conformation and function of the enzyme. Molecular dynamics studies suggest that the loss of interaction between His180 and Tyr89 most likely alters the orientation of Tyr89 side chain, thereby causing the movement of helices α6, α12, α13 and α14 in the vicinity and affecting the tetrameric assembly. We further show that the oligomerization of MtbICL is primarily mediated by the inter subunit interactions, and strengthened by the helix swapping of α12-α13 between adjacent subunits. Furthermore, the enzyme activity is influenced by the interactions between the residues of lid region (P411NSSTTALTGSTEEGQFH428) and the loop region (T391KHQREV397). Mutation of glutamates of the lid region to non homologous residues (E423A or E424A) or basic residues (E423K or E424K) inactivates the enzyme, whereas the activity is not much compromised in case of homologous mutations (E423D or E424D).
Collapse
Affiliation(s)
- Harish Shukla
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, PIN 226 031, India
| | - Vikash Kumar
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, PIN 226 031, India
| | - Amit Kumar Singh
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, PIN 226 031, India
| | - Neha Singh
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, PIN 226 031, India
| | - Md Kashif
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, PIN 226 031, India
| | - Mohammad Imran Siddiqi
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, PIN 226 031, India; Academy of Scientific and Innovative Research, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, PIN 226 031, India
| | - Manju Yasoda Krishnan
- Microbiology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, PIN 226 031, India; Academy of Scientific and Innovative Research, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, PIN 226 031, India
| | - Md Sohail Akhtar
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, PIN 226 031, India; Academy of Scientific and Innovative Research, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, PIN 226 031, India.
| |
Collapse
|
182
|
Karad SC, Purohit VB, Raval DK, Kalaria PN, Avalani JR, Thakor P, Thakkar VR. Green synthesis and pharmacological screening of polyhydroquinoline derivatives bearing a fluorinated 5-aryloxypyrazole nucleus. RSC Adv 2015. [DOI: 10.1039/c5ra00388a] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Green synthesis, biological evaluation and SAR study of polyhydroquinoline scaffold bearing fluorinated 5-aryloxypyrazole nucleus is discussed.
Collapse
Affiliation(s)
- Sharad C. Karad
- Department of Chemistry
- Sardar Patel University
- Vallabh Vidyanagar-388120
- India
| | - Vishal B. Purohit
- Department of Chemistry
- Sardar Patel University
- Vallabh Vidyanagar-388120
- India
| | - Dipak K. Raval
- Department of Chemistry
- Sardar Patel University
- Vallabh Vidyanagar-388120
- India
| | - Piyush N. Kalaria
- Department of Chemistry
- Sardar Patel University
- Vallabh Vidyanagar-388120
- India
| | - Jemin R. Avalani
- Department of Chemistry
- Sardar Patel University
- Vallabh Vidyanagar-388120
- India
| | - Parth Thakor
- B. R. Doshi School of Biosciences
- Sardar Patel Maidan
- Satellite Campus
- Sardar Patel University
- Vallabh Vidyanagar-388120
| | - Vasudev R. Thakkar
- B. R. Doshi School of Biosciences
- Sardar Patel Maidan
- Satellite Campus
- Sardar Patel University
- Vallabh Vidyanagar-388120
| |
Collapse
|
183
|
TOLA HH, TOL A, SHOJAEIZADEH D, GARMAROUDI G. Tuberculosis Treatment Non-Adherence and Lost to Follow Up among TB Patients with or without HIV in Developing Countries: A Systematic Review. IRANIAN JOURNAL OF PUBLIC HEALTH 2015; 44:1-11. [PMID: 26060770 PMCID: PMC4449995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 11/11/2014] [Indexed: 11/05/2022]
Abstract
This systematic review intended to combine factors associated with tuberculosis treatment non-adherence and lost to follow up among TB patients with/without HIV in developing countries. Comprehensive remote electronic databases (MEDLINE, (PMC, Pub Med Central), Google scholar and Web of science) search was conducted using the following keywords: Tuberculosis, treatment, compliance, adherence, default, behavioural factors and socioeconomic factors. All types of studies intended to assess TB treatment non-adherence and lost to follow up in developing countries among adult TB patient from 2008 to data extraction date were included. Twenty-six original and one-reviewed articles, which meet inclusion criteria, were reviewed. TB treatment non-adherence and lost to follow up were continued across developing countries. The main factors associated with TB treatment non-adherence and lost to follow up were socioeconomic factors: lack of transportation cost, lack of social support, and patients-health care worker poor communication. Behavioural factors were Feeling better after few weeks of treatments, tobacco and alcohol use, knowledge deficit about duration of treatment and consequences of non-adherence and lost to follow up. TB treatment non-adherence and lost to follow up were continued across developing countries throughout the publication years of reviewed articles. Numerous, socioeconomic and behavioural factors were influencing TB treatment adherence and lost to follow up. Therefore, well understanding and minimizing of the effect of these associated factors is very important to enhance treatment adherence and follow up completion in developing countries.
Collapse
|
184
|
Ruthenium(II) complexes with hydroxypyridinecarboxylates: Screening potential metallodrugs against Mycobacterium tuberculosis. Polyhedron 2015. [DOI: 10.1016/j.poly.2014.08.057] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
185
|
Zhao P, Li D, Yang F, Ma Y, Wang T, Duan S, Shen H, Cai Q, Wu D, Yang X, Wang S. In vitro and in vivo drug release behavior and osteogenic potential of a composite scaffold based on poly(ε-caprolactone)-block-poly(lactic-co-glycolic acid) and β-tricalcium phosphate. J Mater Chem B 2015; 3:6885-6896. [DOI: 10.1039/c5tb00946d] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To cure serious bone tuberculosis, a novel long-term drug delivery system was designed and prepared to satisfy the needs of both bone regeneration and antituberculous drug therapy.
Collapse
|
186
|
Gowthaman U, Mushtaq K, Tan AC, Rai PK, Jackson DC, Agrewala JN. Challenges and solutions for a rational vaccine design for TB-endemic regions. Crit Rev Microbiol 2015; 41:389-98. [PMID: 24495096 DOI: 10.3109/1040841x.2013.859125] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Vaccines have been successful for global eradication or control of dreaded diseases such as smallpox, diphtheria, tetanus, yellow fever, whooping cough, polio, and measles. Unfortunately, this success has not been achieved for controlling tuberculosis (TB) worldwide. Bacillus Calmette Guérin (BCG) is the only available vaccine against TB. Paradoxically, BCG has deciphered success in the Western world but has failed in TB-endemic areas. In this article, we highlight and discuss the aspects of immunity responsible for controlling Mycobacterium tuberculosis infection and factors responsible for the failure of BCG in TB-endemic countries. In addition, we also suggest strategies that contribute toward the development of successful vaccine in protecting populations where BCG has failed.
Collapse
|
187
|
Subfamily-specific adaptations in the structures of two penicillin-binding proteins from Mycobacterium tuberculosis. PLoS One 2014; 9:e116249. [PMID: 25551456 PMCID: PMC4281109 DOI: 10.1371/journal.pone.0116249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 12/05/2014] [Indexed: 11/19/2022] Open
Abstract
Beta-lactam antibiotics target penicillin-binding proteins including several enzyme classes essential for bacterial cell-wall homeostasis. To better understand the functional and inhibitor-binding specificities of penicillin-binding proteins from the pathogen, Mycobacterium tuberculosis, we carried out structural and phylogenetic analysis of two predicted D,D-carboxypeptidases, Rv2911 and Rv3330. Optimization of Rv2911 for crystallization using directed evolution and the GFP folding reporter method yielded a soluble quadruple mutant. Structures of optimized Rv2911 bound to phenylmethylsulfonyl fluoride and Rv3330 bound to meropenem show that, in contrast to the nonspecific inhibitor, meropenem forms an extended interaction with the enzyme along a conserved surface. Phylogenetic analysis shows that Rv2911 and Rv3330 belong to different clades that emerged in Actinobacteria and are not represented in model organisms such as Escherichia coli and Bacillus subtilis. Clade-specific adaptations allow these enzymes to fulfill distinct physiological roles despite strict conservation of core catalytic residues. The characteristic differences include potential protein-protein interaction surfaces and specificity-determining residues surrounding the catalytic site. Overall, these structural insights lay the groundwork to develop improved beta-lactam therapeutics for tuberculosis.
Collapse
|
188
|
Orme IM, Basaraba RJ. The formation of the granuloma in tuberculosis infection. Semin Immunol 2014; 26:601-9. [DOI: 10.1016/j.smim.2014.09.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 09/11/2014] [Accepted: 09/15/2014] [Indexed: 12/13/2022]
|
189
|
Dorhoi A, Kaufmann SH. Perspectives on host adaptation in response to Mycobacterium tuberculosis: Modulation of inflammation. Semin Immunol 2014; 26:533-42. [DOI: 10.1016/j.smim.2014.10.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 09/30/2014] [Accepted: 10/01/2014] [Indexed: 12/11/2022]
|
190
|
Green J, Rolfe MD, Smith LJ. Transcriptional regulation of bacterial virulence gene expression by molecular oxygen and nitric oxide. Virulence 2014; 5:794-809. [PMID: 25603427 PMCID: PMC4601167 DOI: 10.4161/viru.27794] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Molecular oxygen (O2) and nitric oxide (NO) are diatomic gases that play major roles in infection. The host innate immune system generates reactive oxygen species and NO as bacteriocidal agents and both require O2 for their production. Furthermore, the ability to adapt to changes in O2 availability is crucial for many bacterial pathogens, as many niches within a host are hypoxic. Pathogenic bacteria have evolved transcriptional regulatory systems that perceive these gases and respond by reprogramming gene expression. Direct sensors possess iron-containing co-factors (iron–sulfur clusters, mononuclear iron, heme) or reactive cysteine thiols that react with O2 and/or NO. Indirect sensors perceive the physiological effects of O2 starvation. Thus, O2 and NO act as environmental cues that trigger the coordinated expression of virulence genes and metabolic adaptations necessary for survival within a host. Here, the mechanisms of signal perception by key O2- and NO-responsive bacterial transcription factors and the effects on virulence gene expression are reviewed, followed by consideration of these aspects of gene regulation in two major pathogens, Staphylococcus aureus and Mycobacterium tuberculosis.
Collapse
Key Words
- AIP, autoinducer peptide
- Arc, Aerobic respiratory control
- FNR
- FNR, fumarate nitrate reduction regulator
- GAF, cGMP-specific phosphodiesterase-adenylyl cyclase-FhlA domain
- Isc, iron–sulfur cluster biosynthesis machinery
- Mycobacterium tuberculosis
- NOX, NADPH oxidase
- PAS, Per-Amt-Sim domain
- RNS, reactive nitrogen species
- ROS, reactive oxygen species
- Staphylococcus aureus
- TB, tuberculosis
- WhiB-like proteins
- iNOS, inducible nitric oxide synthase
- iron–sulfur cluster
- nitric oxide sensors
- oxygen sensors
Collapse
Affiliation(s)
- Jeffrey Green
- a Krebs Institute; Molecular Biology & Biotechnology; University of Sheffield ; Western Bank , Sheffield , UK
| | | | | |
Collapse
|
191
|
Francis RJ, Butler RE, Stewart GR. Mycobacterium tuberculosis ESAT-6 is a leukocidin causing Ca2+ influx, necrosis and neutrophil extracellular trap formation. Cell Death Dis 2014; 5:e1474. [PMID: 25321481 PMCID: PMC4237235 DOI: 10.1038/cddis.2014.394] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 08/08/2014] [Accepted: 08/14/2014] [Indexed: 11/08/2022]
Abstract
Mycobacterium tuberculosis infection generates pulmonary granulomas that consist of a caseous, necrotic core surrounded by an ordered arrangement of macrophages, neutrophils and T cells. This inflammatory pathology is essential for disease transmission and M. tuberculosis has evolved to stimulate inflammatory granuloma development while simultaneously avoiding destruction by the attracted phagocytes. The most abundant phagocyte in active necrotic granulomas is the neutrophil. Here we show that the ESAT-6 protein secreted by the ESX-1 type VII secretion system causes necrosis of the neutrophils. ESAT-6 induced an intracellular Ca(2+) overload followed by necrosis of phosphatidylserine externalised neutrophils. This necrosis was dependent upon the Ca(2+) activated protease calpain, as pharmacologic inhibition prevented this secondary necrosis. We also observed that the ESAT-6 induced increase in intracellular Ca(2+), stimulated the production of neutrophil extracellular traps characterised by extruded DNA and myeloperoxidase. Thus we conclude that ESAT-6 has a leukocidin function, which may facilitate bacterial avoidance of the antimicrobial action of the neutrophil while contributing to the maintenance of inflammation and necrotic pathology necessary for granuloma formation and TB transmission.
Collapse
Affiliation(s)
- R J Francis
- Department of Microbial and Cellular Sciences and Bioimaging and Flow Cytometry Core Facility, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - R E Butler
- Department of Microbial and Cellular Sciences and Bioimaging and Flow Cytometry Core Facility, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - G R Stewart
- Department of Microbial and Cellular Sciences and Bioimaging and Flow Cytometry Core Facility, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| |
Collapse
|
192
|
Wang J, Huang C, Wu M, Zhong Q, Yang K, Li M, Zhan X, Wen J, Zhou L, Huang X. MRP8/14 induces autophagy to eliminate intracellular Mycobacterium bovis BCG. J Infect 2014; 70:415-26. [PMID: 25312864 DOI: 10.1016/j.jinf.2014.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 08/30/2014] [Accepted: 09/15/2014] [Indexed: 01/30/2023]
Abstract
OBJECTIVE To explore the role of myeloid-related protein 8/14 in mycobacterial infection. METHODS The mRNA and protein expression levels of MRP8 or MRP14 were measured by real-time PCR and flow cytometry, respectively. Role of MRP8/14 was tested by overexpression or RNA interference assays. Flow cytometry and colony forming unit were used to test the phagocytosis and the survival of intracellular Mycobacterium bovis BCG (BCG), respectively. Autophagy mediated by MRP8/14 was detected by Western blot and immunofluorescence. The colocalization of BCG phagosomes with autophagosomes or lysosomes was by detected by confocal microscopy. ROS production was detected by flow cytometry. RESULTS MRP8/14 expressions were up-regulated in human monocytic THP1 cells and primary macrophages after mycobacterial challenge. Silencing of MRP8/14 suppressed bacterial killing, but had no influence on the phagocytosis of BCG. Importantly, silencing MRP8/14 decreased autophagy and BCG phagosome maturation in THP1-derived macrophages, thereby increasing the BCG survival. Additionally, we demonstrated that MRP8/14 promoted autophagy in a ROS-dependent manner. CONCLUSIONS The present study revealed a novel role of MRP8/14 in the autophagy-mediated elimination of intracellular BCG by promoting ROS generation, which may provide a promising therapeutic target for tuberculosis and other intracellular bacterial infectious diseases.
Collapse
Affiliation(s)
- Jinli Wang
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Department of Laboratory Medicine, Guangzhou First Municipal People's Hospital, Affiliated Hospital of Guangzhou Medical University, Guangzhou 510500, China
| | - Chunyu Huang
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, China
| | - Minhao Wu
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Qiu Zhong
- Center for Tuberculosis Control of Guangdong Province, Guangzhou 510630, China
| | - Kun Yang
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Miao Li
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Xiaoxia Zhan
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Jinsheng Wen
- Department of Microbiology and Immunology, Wenzhou Medical University, Wenzhou 325035, China
| | - Lin Zhou
- Center for Tuberculosis Control of Guangdong Province, Guangzhou 510630, China.
| | - Xi Huang
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Department of Microbiology and Immunology, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
193
|
BoseDasgupta S, Pieters J. Striking the Right Balance Determines TB or Not TB. Front Immunol 2014; 5:455. [PMID: 25339950 PMCID: PMC4189424 DOI: 10.3389/fimmu.2014.00455] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/06/2014] [Indexed: 12/11/2022] Open
Abstract
Mycobacterium tuberculosis continues to be one of the most successful pathogens on earth. Upon inhalation of M. tuberculosis by a healthy individual, the host immune system will attempt to eliminate these pathogens using a combination of immune defense strategies. These include the recruitment of macrophages and other phagocytes to the site of infection, production of cytokines that enhance the microbicidal capacity of the macrophages, as well as the activation of distinct subsets of leukocytes that work in concert to fight the infection. However, being as successful as it is, M. tuberculosis has evolved numerous strategies to subvert host immunity at virtual every level. As a consequence, one third of the world inhabitants carry M. tuberculosis, and tuberculosis continuous to cause disease in more than 8 million people with deadly consequences in well over 1 million patients each year. In this review, we discuss several of the strategies that M. tuberculosis employs to circumvent host immunity, as well as describe some of the mechanisms that the host uses to counter such subversive strategies. As for many other infectious diseases, the ultimate outcome is usually defined by the relative strength of the virulence strategies employed by the tubercle bacillus versus the arsenal of immune defense mechanisms of the infected host.
Collapse
Affiliation(s)
| | - Jean Pieters
- Biozentrum, University of Basel , Basel , Switzerland
| |
Collapse
|
194
|
Garg NK, Dwivedi P, Jain A, Tyagi S, Sahu T, Tyagi RK. Development of novel carrier(s) mediated tuberculosis vaccine: more than a tour de force. Eur J Pharm Sci 2014; 62:227-242. [PMID: 24909731 DOI: 10.1016/j.ejps.2014.05.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 04/05/2014] [Accepted: 05/28/2014] [Indexed: 02/07/2023]
Abstract
Despite worldwide availability of the vaccines against most of the infectious diseases, BCG and various programs such as Directly Observed Treatment Short course (DOTS) to prevent tuberculosis still remains one of the most deadly forms of the disease affecting millions of people globally. The evolution of multi drug resistant strains (MDR) has increased the complexity further. Although currently available marketed BCG vaccine has shown sufficient protection against childhood tuberculosis, it has failed to prevent the most common form of disease i.e., pulmonary tuberculosis in adults. However, various vaccine candidates have already entered phase I clinical trials and have shown promising outcomes. The most prominent amongst them is the heterologous prime-boost approach, which shows a great promise towards designing and development of a new efficacious tuberculosis vaccine. It has also been shown that the use of various viral and non-viral vectors as carriers for the potential vaccine candidates will further boost their effect on subsequent immunization. In this review, we briefly summarize the potential of a few novel nano-carriers for developing effective vaccination strategies against tuberculosis.
Collapse
Affiliation(s)
- Neeraj K Garg
- Drug Delivery Research Group, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies, Panjab University, 160 014 Chandigarh, India; Department of Pharmaceutical Sciences, Dr. H.S. Gour University, Sagar 470 003, MP, India.
| | - Priya Dwivedi
- Department of Biotechnology, TRS College, Rewa 486001, MP, India
| | - Ashay Jain
- Drug Delivery Research Group, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies, Panjab University, 160 014 Chandigarh, India; Department of Pharmaceutical Sciences, Dr. H.S. Gour University, Sagar 470 003, MP, India
| | - Shikha Tyagi
- Department of Biotechnology, IMS Engineering College, Ghaziabad, UP Technical University, UP, India
| | - Tejram Sahu
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, TW3/3W15, 12735 Twinbrook Pkwy, Rockville, MD, USA
| | - Rajeev K Tyagi
- Department of Periodontics, College of Dental Medicine, Georgia Regents University, Augusta, GA, USA.
| |
Collapse
|
195
|
Vickery CR, Kosa NM, Casavant EP, Duan S, Noel JP, Burkart MD. Structure, biochemistry, and inhibition of essential 4'-phosphopantetheinyl transferases from two species of Mycobacteria. ACS Chem Biol 2014; 9:1939-44. [PMID: 24963544 PMCID: PMC4168790 DOI: 10.1021/cb500263p] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
![]()
4′-Phosphopantetheinyl
transferases (PPTase) post-translationally
modify carrier proteins with a phosphopantetheine moiety, an essential
reaction in all three domains of life. In the bacterial genus Mycobacteria, the Sfp-type PPTase activates pathways necessary
for the biosynthesis of cell wall components and small molecule virulence
factors. We solved the X-ray crystal structures and biochemically
characterized the Sfp-type PPTases from two of the most prevalent
Mycobacterial pathogens, PptT of M. tuberculosis and
MuPPT of M. ulcerans. Structural analyses reveal
significant differences in cofactor binding and active site composition
when compared to previously characterized Sfp-type PPTases. Functional
analyses including the efficacy of Sfp-type PPTase-specific inhibitors
also suggest that the Mycobacterial Sfp-type PPTases can serve as
therapeutic targets against Mycobacterial infections.
Collapse
Affiliation(s)
- Christopher R. Vickery
- Department
of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
- Jack
Skirball Center for Chemical Biology and Proteomics, The Salk Institute, 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Nicolas M. Kosa
- Department
of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Ellen P. Casavant
- Department
of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Shiteng Duan
- Department
of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Joseph P. Noel
- Howard Hughes Medical Institute, 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
- Jack
Skirball Center for Chemical Biology and Proteomics, The Salk Institute, 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Michael D. Burkart
- Department
of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| |
Collapse
|
196
|
Sukumar N, Tan S, Aldridge BB, Russell DG. Exploitation of Mycobacterium tuberculosis reporter strains to probe the impact of vaccination at sites of infection. PLoS Pathog 2014; 10:e1004394. [PMID: 25233380 PMCID: PMC4169503 DOI: 10.1371/journal.ppat.1004394] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 08/12/2014] [Indexed: 11/19/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) remains a major public health problem, with an effective vaccine continuing to prove elusive. Progress in vaccination strategies has been hampered by a lack of appreciation of the bacterium's response to dynamic changes in the host immune environment. Here, we utilize reporter Mtb strains that respond to specific host immune stresses such as hypoxia and nitric oxide (hspX'::GFP), and phagosomal maturation (rv2390c'::GFP), to investigate vaccine-induced alterations in the environmental niche during experimental murine infections. While vaccination undoubtedly decreased bacterial burden, we found that it also appeared to accelerate Mtb's adoption of a phenotype better equipped to survive in its host. We subsequently utilized a novel replication reporter strain of Mtb to demonstrate that, in addition to these alterations in host stress response, there is a decreased percentage of actively replicating Mtb in vaccinated hosts. This observation was supported by the differential sensitivity of recovered bacteria to the front-line drug isoniazid. Our study documents the natural history of the impact that vaccination has on Mtb's physiology and replication and highlights the value of reporter Mtb strains for probing heterogeneous Mtb populations in the context of a complex, whole animal model.
Collapse
Affiliation(s)
- Neelima Sukumar
- Cornell University, College of Veterinary Medicine, Department of Microbiology and Immunology, Ithaca, New York, United States of America
| | - Shumin Tan
- Cornell University, College of Veterinary Medicine, Department of Microbiology and Immunology, Ithaca, New York, United States of America
| | - Bree B. Aldridge
- Tufts University School of Medicine, Department of Molecular Biology and Microbiology, Boston, Massachusetts, United States of America
| | - David G. Russell
- Cornell University, College of Veterinary Medicine, Department of Microbiology and Immunology, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
197
|
Govender VS, Ramsugit S, Pillay M. Mycobacterium tuberculosis adhesins: potential biomarkers as anti-tuberculosis therapeutic and diagnostic targets. Microbiology (Reading) 2014; 160:1821-1831. [DOI: 10.1099/mic.0.082206-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Adhesion to host cells is a precursor to host colonization and evasion of the host immune response. Conversely, it triggers the induction of the immune response, a process vital to the host’s defence against infection. Adhesins are microbial cell surface molecules or structures that mediate the attachment of the microbe to host cells and thus the host–pathogen interaction. They also play a crucial role in bacterial aggregation and biofilm formation. In this review, we discuss the role of adhesins in the pathogenesis of the aetiological agent of tuberculosis, Mycobacterium tuberculosis. We also provide insight into the structure and characteristics of some of the characterized and putative M. tuberculosis adhesins. Finally, we examine the potential of adhesins as targets for the development of tuberculosis control strategies.
Collapse
Affiliation(s)
- Viveshree S. Govender
- Medical Microbiology and Infection Control, University of KwaZulu-Natal, Durban, South Africa
| | - Saiyur Ramsugit
- Medical Microbiology and Infection Control, University of KwaZulu-Natal, Durban, South Africa
| | - Manormoney Pillay
- Medical Microbiology and Infection Control, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
198
|
Edagwa BJ, Guo D, Puligujja P, Chen H, McMillan J, Liu X, Gendelman HE, Narayanasamy P. Long-acting antituberculous therapeutic nanoparticles target macrophage endosomes. FASEB J 2014; 28:5071-82. [PMID: 25122556 PMCID: PMC4232285 DOI: 10.1096/fj.14-255786] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Eradication of Mycobacterium tuberculosis (MTB) infection requires daily administration of combinations of rifampin (RIF), isoniazid [isonicotinylhydrazine (INH)], pyrazinamide, and ethambutol, among other drug therapies. To facilitate and optimize MTB therapeutic selections, a mononuclear phagocyte (MP; monocyte, macrophage, and dendritic cell)-targeted drug delivery strategy was developed. Long-acting nanoformulations of RIF and an INH derivative, pentenyl-INH (INHP), were prepared, and their physicochemical properties were evaluated. This included the evaluation of MP particle uptake and retention, cell viability, and antimicrobial efficacy. Drug levels reached 6 μg/10(6) cells in human monocyte-derived macrophages (MDMs) for nanoparticle treatments compared with 0.1 μg/10(6) cells for native drugs. High RIF and INHP levels were retained in MDM for >15 d following nanoparticle loading. Rapid loss of native drugs was observed in cells and culture fluids within 24 h. Antimicrobial activities were determined against Mycobacterium smegmatis (M. smegmatis). Coadministration of nanoformulated RIF and INHP provided a 6-fold increase in therapeutic efficacy compared with equivalent concentrations of native drugs. Notably, nanoformulated RIF and INHP were found to be localized in recycling and late MDM endosomal compartments. These were the same compartments that contained the pathogen. Our results demonstrate the potential of antimicrobial nanomedicines to simplify MTB drug regimens.
Collapse
Affiliation(s)
- Benson J Edagwa
- Department of Pharmacology and Experimental Neuroscience, and
| | - Dongwei Guo
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA; and
| | - Pavan Puligujja
- Department of Pharmacology and Experimental Neuroscience, and
| | - Han Chen
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | | | - Xinming Liu
- Department of Pharmacology and Experimental Neuroscience, and
| | | | | |
Collapse
|
199
|
Nair V, Okello M, Mishra S, Mirsalis J, O'Loughlin K, Zhong Y. Pharmacokinetics and dose-range finding toxicity of a novel anti-HIV active integrase inhibitor. Antiviral Res 2014; 108:25-9. [PMID: 24821255 PMCID: PMC4101043 DOI: 10.1016/j.antiviral.2014.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 04/30/2014] [Accepted: 05/02/2014] [Indexed: 12/27/2022]
Abstract
Integration of viral DNA into human chromosomal DNA catalyzed by HIV integrase represents the "point of no return" in HIV infection. For this reason, HIV integrase is considered a crucial target in the development of new anti-HIV therapeutic agents. We have discovered a novel HIV integrase inhibitor 1, that exhibits potent antiviral activity and a favorable metabolism profile. This paper reports on the pharmacokinetics and toxicokinetics of compound 1 and the relevance of these findings with respect to further development of this integrase-targeted antiviral agent. Oral administration of compound 1 in Sprague Dawley rats revealed rapid absorption. Drug exposure increased with increasing drug concentration, indicative of appropriate dose-dependence correlation. Compound 1 exhibited suitable plasma half-life, extensive extravascular distribution and acceptable bioavailability. Toxicity studies revealed no compound-related clinical pathology findings. There were no changes in erythropoietic, white blood cell or platelet parameters in male and female rats. There was no test-article related change in other clinical chemistry parameters. In addition, there were no detectable levels of bilirubin in the urine and there were no treatment-related effects on urobilinogen or other urinalysis parameters. The preclinical studies also revealed that the no observed adverse effect level and the maximum tolerated dose were both high (>500mg/kg/day). The broad and significant antiviral activity and favorable metabolism profile of this integrase inhibitor, when combined with the in vivo pharmacokinetic and toxicokinetic data and their pharmacological relevance, provide compelling and critical support for its further development as an anti-HIV therapeutic agent.
Collapse
Affiliation(s)
- Vasu Nair
- Center for Drug Discovery and the College of Pharmacy University of Georgia, Athens, GA 30602, USA.
| | - Maurice Okello
- Center for Drug Discovery and the College of Pharmacy University of Georgia, Athens, GA 30602, USA
| | - Sanjay Mishra
- Center for Drug Discovery and the College of Pharmacy University of Georgia, Athens, GA 30602, USA
| | - Jon Mirsalis
- Biosciences Division, SRI International, Menlo Park, CA 94025, USA
| | | | - Yu Zhong
- Biosciences Division, SRI International, Menlo Park, CA 94025, USA
| |
Collapse
|
200
|
Mehrotra P, Jamwal SV, Saquib N, Sinha N, Siddiqui Z, Manivel V, Chatterjee S, Rao KVS. Pathogenicity of Mycobacterium tuberculosis is expressed by regulating metabolic thresholds of the host macrophage. PLoS Pathog 2014; 10:e1004265. [PMID: 25058590 PMCID: PMC4110042 DOI: 10.1371/journal.ppat.1004265] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 06/06/2014] [Indexed: 11/18/2022] Open
Abstract
The success of Mycobacterium tuberculosis as a pathogen derives from its facile adaptation to the intracellular milieu of human macrophages. To explore this process, we asked whether adaptation also required interference with the metabolic machinery of the host cell. Temporal profiling of the metabolic flux, in cells infected with differently virulent mycobacterial strains, confirmed that this was indeed the case. Subsequent analysis identified the core subset of host reactions that were targeted. It also elucidated that the goal of regulation was to integrate pathways facilitating macrophage survival, with those promoting mycobacterial sustenance. Intriguingly, this synthesis then provided an axis where both host- and pathogen-derived factors converged to define determinants of pathogenicity. Consequently, whereas the requirement for macrophage survival sensitized TB susceptibility to the glycemic status of the individual, mediation by pathogen ensured that the virulence properties of the infecting strain also contributed towards the resulting pathology. Mycobacterium tuberculosis (Mtb) is a highly successful human pathogen, representing the leading bacterial cause of death worldwide. Mtb infects macrophages and it adapts to the hostile intracellular milieu of this cell by exploiting the plasticity of its central carbon metabolism machinery. While several studies have detailed the bacterial adaptations that accompany infection, it is still unclear whether this process also involves engagement with host metabolic pathways. We therefore profiled the kinetic flux of host cell metabolites in macrophages that were infected with differently virulent Mtb strains. Interestingly, we found that Mtb pathogenicity was indeed intimately linked to its capacity to regulate host cell metabolism. A unique subset of host pathways was targeted so as to integrate the glycolytic threshold governing macrophage viability with mechanisms ensuring intracellular bacterial survival. Perturbation of macrophage glycolytic flux was enforced through pathogen-induced enhancement in glucose uptake, which in turn was also influenced by the extracellular glucose concentration. This observation rationalizes the increased susceptibility of diabetic individuals to TB infection Interestingly, Mtb strains also differed in their capacities to stimulate macrophage glucose uptake. Consequently, the resulting pathology is likely dictated both by the individual's glycemic status, and the nature of the infecting strain.
Collapse
Affiliation(s)
- Parul Mehrotra
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Shilpa V. Jamwal
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Najmuddin Saquib
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Neeraj Sinha
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Zaved Siddiqui
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Venkatasamy Manivel
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Samrat Chatterjee
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Kanury V. S. Rao
- Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- * E-mail:
| |
Collapse
|