151
|
Zheng X, Av-Gay Y. New Era of TB Drug Discovery and Its Impact on Disease Management. CURRENT TREATMENT OPTIONS IN INFECTIOUS DISEASES 2016. [DOI: 10.1007/s40506-016-0098-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
152
|
Abstract
One quarter of all deaths worldwide each year result from infectious diseases caused by microbial pathogens. Pathogens infect and cause disease by producing virulence factors that target host cell molecules. Studying how virulence factors target host cells has revealed fundamental principles of cell biology. These include important advances in our understanding of the cytoskeleton, organelles and membrane-trafficking intermediates, signal transduction pathways, cell cycle regulators, the organelle/protein recycling machinery, and cell-death pathways. Such studies have also revealed cellular pathways crucial for the immune response. Discoveries from basic research on the cell biology of pathogenesis are actively being translated into the development of host-targeted therapies to treat infectious diseases. Thus there are many reasons for cell biologists to incorporate the study of microbial pathogens into their research programs.
Collapse
Affiliation(s)
- Matthew D Welch
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
153
|
Whisson SC, Boevink PC, Wang S, Birch PR. The cell biology of late blight disease. Curr Opin Microbiol 2016; 34:127-135. [PMID: 27723513 PMCID: PMC5340842 DOI: 10.1016/j.mib.2016.09.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/01/2016] [Accepted: 09/08/2016] [Indexed: 11/12/2022]
Abstract
The Phytophthora haustorium is a major site of secretion during infection. The host endocytic cycle contributes to biogenesis of the Extra-Haustorial Membrane. RXLR effectors manipulate host processes at diverse subcellular locations. They directly manipulate the activity or location of immune proteins. They also promote the activity of endogenous negative regulators of immunity.
Late blight, caused by the oomycete Phytophthora infestans, is a major global disease of potato and tomato. Cell biology is teaching us much about the developmental stages associated with infection, especially the haustorium, which is a site of intimate interaction and molecular exchange between pathogen and host. Recent observations suggest a role for the plant endocytic cycle in specific recruitment of host proteins to the Extra-Haustorial Membrane, emphasising the unique nature of this membrane compartment. In addition, there has been a strong focus on the activities of RXLR effectors, which are delivered into plant cells to modulate and manipulate host processes. RXLR effectors interact directly with diverse plant proteins at a range of subcellular locations to promote disease.
Collapse
Affiliation(s)
- Stephen C Whisson
- Cell and Molecular Sciences, James Hutton Institute, Errol Road, Invergowrie, Dundee DD2 5DA, UK
| | - Petra C Boevink
- Cell and Molecular Sciences, James Hutton Institute, Errol Road, Invergowrie, Dundee DD2 5DA, UK
| | - Shumei Wang
- Division of Plant Sciences, University of Dundee, James Hutton Institute, Invergowrie, Dundee DD2 5DA, UK
| | - Paul Rj Birch
- Cell and Molecular Sciences, James Hutton Institute, Errol Road, Invergowrie, Dundee DD2 5DA, UK; Division of Plant Sciences, University of Dundee, James Hutton Institute, Invergowrie, Dundee DD2 5DA, UK.
| |
Collapse
|
154
|
Singh V, Mizrahi V. Identification and validation of novel drug targets in Mycobacterium tuberculosis. Drug Discov Today 2016; 22:503-509. [PMID: 27649943 DOI: 10.1016/j.drudis.2016.09.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/28/2016] [Accepted: 09/12/2016] [Indexed: 10/21/2022]
Abstract
Tuberculosis (TB) is a global epidemic associated increasingly with resistance to first- and second-line antitubercular drugs. The magnitude of this global health threat underscores the urgent need to discover new antimycobacterial agents that have novel mechanisms of action (MOA). In this review, we highlight some of the key advances that have enabled the strengths of target-led and phenotypic approaches to TB drug discovery to be harnessed both independently and in combination. Critically, these promise to fuel the front-end of the TB drug pipeline with new, pharmacologically validated drug targets together with lead compounds that act on these targets.
Collapse
Affiliation(s)
- Vinayak Singh
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease and Molecular Medicine & Department of Pathology, University of Cape Town, Anzio Road, Observatory 7925, South Africa.
| | - Valerie Mizrahi
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease and Molecular Medicine & Department of Pathology, University of Cape Town, Anzio Road, Observatory 7925, South Africa
| |
Collapse
|
155
|
Bis-biguanide dihydrochloride inhibits intracellular replication of M. tuberculosis and controls infection in mice. Sci Rep 2016; 6:32725. [PMID: 27601302 PMCID: PMC5013693 DOI: 10.1038/srep32725] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/15/2016] [Indexed: 12/18/2022] Open
Abstract
While there is an urgent need to develop new and effective drugs for treatment of tuberculosis (TB) and multi-drug resistant TB (MDR-TB), repurposing FDA (U.S. Food and Drug Administration) -approved drugs for development of anti-TB agents may decrease time and effort from bench to bedside. Here, we employed host cell-based high throughput screening (HTS) assay to screen and characterize FDA-approved, off-patent library drugs for anti-Mycobacterium tuberculosis (MTB) activities. The cell-based HTS allowed us to identify an anti-cancer drug of bis-biguanide dihydrochloride (BBD) as potent anti-mycobacteria agent. Further characterization showed that BBD could inhibit intracellular and extracellular growth of M. smegmatis and slow-growing M. bovis BCG. BBD also potently inhibited replication of clinically-isolated MTB and MDR-TB strains. The proof-of-concept study showed that BBD treatment of MTB-infected mice could significantly decrease CFU counts in the lung and spleen. Notably, comparative evaluation showed that MTB CFU counts in BBD-treated mice were lower than those in rifampicin-treated mice. No apparent BBD side effects were found in BBD-treated mice. Thus, our findings support further studies to develop BBD as a new and effective drug against TB and MDR-TB.
Collapse
|
156
|
A Rapid Method for Quantifying Viable Mycobacterium avium subsp. paratuberculosis in Cellular Infection Assays. Appl Environ Microbiol 2016; 82:5553-62. [PMID: 27371585 DOI: 10.1128/aem.01668-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 06/27/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Determining the viability of bacteria is a key outcome of in vitro cellular infection assays. Currently, this is done by culture, which is problematic for fastidious slow-growing bacteria such as Mycobacterium avium subsp. paratuberculosis, where it can take up to 4 months to confirm growth. This study aimed to identify an assay that can rapidly quantify the number of viable M. avium subsp. paratuberculosis cells in a cellular sample. Three commercially available bacterial viability assays along with a modified liquid culture method coupled with high-throughput quantitative PCR growth detection were assessed. Criteria for assessment included the ability of each assay to differentiate live and dead M. avium subsp. paratuberculosis organisms and their accuracy at low bacterial concentrations. Using the culture-based method, M. avium subsp. paratuberculosis growth was reliably detected and quantified within 2 weeks. There was a strong linear association between the 2-week growth rate and the initial inoculum concentration. The number of viable M. avium subsp. paratuberculosis cells in an unknown sample was quantified based on the growth rate, by using growth standards. In contrast, none of the commercially available viability assays were suitable for use with samples from in vitro cellular infection assays. IMPORTANCE Rapid quantification of the viability of Mycobacterium avium subsp. paratuberculosis in samples from in vitro cellular infection assays is important, as it allows these assays to be carried out on a large scale. In vitro cellular infection assays can function as a preliminary screening tool, for vaccine development or antimicrobial screening, and also to extend findings derived from experimental animal trials. Currently, by using culture, it takes up to 4 months to obtain quantifiable results regarding M. avium subsp. paratuberculosis viability after an in vitro infection assay; however, with the quantitative PCR and liquid culture method developed, reliable results can be obtained at 2 weeks. This method will be important for vaccine and antimicrobial screening work, as it will allow a greater number of candidates to be screened in the same amount of time, which will increase the likelihood that a favorable candidate will be found to be subjected to further testing.
Collapse
|
157
|
Abstract
The family members Batf, Batf2 and Batf3 belong to a class of transcription factors containing basic leucine zipper domains that regulate various immunological functions and control the development and differentiation of immune cells. Functional studies by others demonstrated a predominant role for Batf in controlling Th2 cell functions and lineage development of T lymphocytes as well as a critical role of Batf, Batf2 and Batf3 in CD8α+dendritic cell development. Moreover, Batf family member expression was measured in a vast collection of mouse and human cell types by cap analysis gene expression (CAGE), a recent developed sequencing technology, showing reasonable expression spectrum in immune cells consistent with previously published expression profiles. Batf and Batf3 were highly expressed in lymphocytes and the earlier moderately expressed in myeloid lineages. Batf2 was predominantly expressed in monocytes/macrophages. Functional studies in mice demonstrated that Batf2 has a central role in macrophage activation by regulating inflammatory responses during lipopolysaccharides stimulation and mycobacterial infection. Hence, Batf2 could be used as a biomarker and a potential host directed drug target in tuberculosis. Moreover, Batf2 act as a tumor suppressor gene and augmenting Batf2 in malignant cells might be an encouraging therapeutic treatment against cancer.
Collapse
|
158
|
Nazimek K, Strobel S, Bryniarski P, Kozlowski M, Filipczak-Bryniarska I, Bryniarski K. The role of macrophages in anti-inflammatory activity of antidepressant drugs. Immunobiology 2016; 222:823-830. [PMID: 27453459 DOI: 10.1016/j.imbio.2016.07.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/12/2016] [Indexed: 10/21/2022]
Abstract
Depression is a common disease influencing patients' quality of life, whose etiology involves complex interactions of environmental, genetic and immunological factors. The latter factors include proinflammatory activation of monocytes and macrophages and increased serum levels of proinflammatory cytokines, altogether formulated as the "macrophage theory of depression". Our current review summarizes the impact of the most commonly used antidepressant drugs on the immune response with special emphasis on the role of macrophages in the clinically observed effects. The anti-inflammatory action of antidepressants mainly results from their direct interaction with immune cells and from changes in the concentration and the relations of neurotransmitters sensed by these cells. The summarized data revealed that Mφs are one of the leading cell populations involved in drug-mediated immune effects that can be observed both in subjects with depression as well as in individuals not suffering from depression. Thus, currently reviewed immunomodulatory effects of the experimental use of different antidepressant drugs suggest the possibility of utilizing them in complex therapeutic strategies dedicated to various inflammatory and immune-mediated diseases. It is worth noting that an excessive inflammatory reaction is also associated with the pathogenesis of various cardiovascular, metabolic and neuro-endocrine diseases. Thus, the inclusion of antidepressants in the complex therapy of these disorders may have beneficial effects through the enhancement of the mood of the patient and alleviation of chronic inflammation. On the other hand, presented data suggest that the influence of chronically used antidepressants on anti-microbial and anti-tumor immunity could also be taken into consideration.
Collapse
Affiliation(s)
- Katarzyna Nazimek
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St, PL 31-121 Krakow, Poland.
| | - Spencer Strobel
- Students' Scientific Society, Department of Immunology, Jagiellonian University Medical College, 18 Czysta St, PL 31-121 Krakow, Poland.
| | - Paweł Bryniarski
- Students' Scientific Society, Department of Immunology, Jagiellonian University Medical College, 18 Czysta St, PL 31-121 Krakow, Poland; Students' Scientific Society, Department of Pain Treatment and Palliative Care, Jagiellonian University Medical College, 10 Sniadeckich St, PL 31-531 Krakow, Poland.
| | - Michael Kozlowski
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St, PL 31-121 Krakow, Poland; Students' Scientific Society, Department of Pain Treatment and Palliative Care, Jagiellonian University Medical College, 10 Sniadeckich St, PL 31-531 Krakow, Poland.
| | - Iwona Filipczak-Bryniarska
- Department of Pain Treatment and Palliative Care, Jagiellonian University Medical College, 10 Sniadeckich St, PL 31-531 Krakow, Poland.
| | - Krzysztof Bryniarski
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St, PL 31-121 Krakow, Poland.
| |
Collapse
|
159
|
Long J, Basu Roy R, Zhang YJ, Antrobus R, Du Y, Smith DL, Weekes MP, Javid B. Plasma Membrane Profiling Reveals Upregulation of ABCA1 by Infected Macrophages Leading to Restriction of Mycobacterial Growth. Front Microbiol 2016; 7:1086. [PMID: 27462310 PMCID: PMC4940386 DOI: 10.3389/fmicb.2016.01086] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 06/29/2016] [Indexed: 01/01/2023] Open
Abstract
The plasma membrane represents a critical interface between the internal and extracellular environments, and harbors multiple proteins key receptors and transporters that play important roles in restriction of intracellular infection. We applied plasma membrane profiling, a technique that combines quantitative mass spectrometry with selective cell surface aminooxy-biotinylation, to Bacille Calmette–Guérin (BCG)-infected THP-1 macrophages. We quantified 559 PM proteins in BCG-infected THP-1 cells. One significantly upregulated cell-surface protein was the cholesterol transporter ABCA1. We showed that ABCA1 was upregulated on the macrophage cell-surface following infection with pathogenic mycobacteria and knockdown of ABCA1 resulted in increased mycobacterial survival within macrophages, suggesting that it may be a novel mycobacterial host-restriction factor.
Collapse
Affiliation(s)
- Jing Long
- Collaboration Innovation Centre for the Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University Beijing, China
| | | | | | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge Cambridge, UK
| | - Yuxian Du
- Collaboration Innovation Centre for the Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University Beijing, China
| | - Duncan L Smith
- Cancer Research UK Manchester Institute, University of Manchester Manchester, UK
| | - Michael P Weekes
- Cambridge Institute for Medical Research, University of Cambridge Cambridge, UK
| | - Babak Javid
- Collaboration Innovation Centre for the Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua UniversityBeijing, China; Harvard TH Chan School of Public Health, BostonMA, USA
| |
Collapse
|
160
|
Mahon RN, Hafner R. Immune Cell Regulatory Pathways Unexplored as Host-Directed Therapeutic Targets for Mycobacterium tuberculosis: An Opportunity to Apply Precision Medicine Innovations to Infectious Diseases. Clin Infect Dis 2016; 61Suppl 3:S200-16. [PMID: 26409283 DOI: 10.1093/cid/civ621] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The lack of novel antimicrobial drugs in development for tuberculosis treatment has provided an impetus for the discovery of adjunctive host-directed therapies (HDTs). Several promising HDT candidates are being evaluated, but major advancement of tuberculosis HDTs will require understanding of the master or "core" cell signaling pathways that control intersecting immunologic and metabolic regulatory mechanisms, collectively described as "immunometabolism." Core regulatory pathways conserved in all eukaryotic cells include poly (ADP-ribose) polymerases (PARPs), sirtuins, AMP-activated protein kinase (AMPK), and mechanistic target of rapamycin (mTOR) signaling. Critical interactions of these signaling pathways with each other and their roles as master regulators of immunometabolic functions will be addressed, as well as how Mycobacterium tuberculosis is already known to influence various other cell signaling pathways interacting with them. Knowledge of these essential mechanisms of cell function regulation has led to breakthrough targeted treatment advances for many diseases, most prominently in oncology. Leveraging these exciting advances in precision medicine for the development of innovative next-generation HDTs may lead to entirely new paradigms for treatment and prevention of tuberculosis and other infectious diseases.
Collapse
Affiliation(s)
- Robert N Mahon
- Division of AIDS-Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Contractor to the National Institute of Allergy and Infectious Diseases, National Institutes of Health
| | - Richard Hafner
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
161
|
O'Connor G, Gleeson LE, Fagan-Murphy A, Cryan SA, O'Sullivan MP, Keane J. Sharpening nature's tools for efficient tuberculosis control: A review of the potential role and development of host-directed therapies and strategies for targeted respiratory delivery. Adv Drug Deliv Rev 2016; 102:33-54. [PMID: 27151307 DOI: 10.1016/j.addr.2016.04.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/04/2016] [Accepted: 04/20/2016] [Indexed: 12/18/2022]
Abstract
Centuries since it was first described, tuberculosis (TB) remains a significant global public health issue. Despite ongoing holistic measures implemented by health authorities and a number of new oral treatments reaching the market, there is still a need for an advanced, efficient TB treatment. An adjunctive, host-directed therapy designed to enhance endogenous pathways and hence compliment current regimens could be the answer. The integration of drug repurposing, including synthetic and naturally occurring compounds, with a targeted drug delivery platform is an attractive development option. In order for a new anti-tubercular treatment to be produced in a timely manner, a multidisciplinary approach should be taken from the outset including stakeholders from academia, the pharmaceutical industry, and regulatory bodies keeping the patient as the key focus. Pre-clinical considerations for the development of a targeted host-directed therapy are discussed here.
Collapse
Affiliation(s)
- Gemma O'Connor
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin and St. James's Hospital, D08 W9RT, Dublin, Ireland.
| | - Laura E Gleeson
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin and St. James's Hospital, D08 W9RT, Dublin, Ireland.
| | - Aidan Fagan-Murphy
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; SFI Centre for Research in Medical Devices (CURAM), Dublin 2, Ireland.
| | - Sally-Ann Cryan
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland; SFI Centre for Research in Medical Devices (CURAM), Dublin 2, Ireland.
| | - Mary P O'Sullivan
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin and St. James's Hospital, D08 W9RT, Dublin, Ireland.
| | - Joseph Keane
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin and St. James's Hospital, D08 W9RT, Dublin, Ireland.
| |
Collapse
|
162
|
Goodman A, Carpenter AE. High-Throughput, Automated Image Processing for Large-Scale Fluorescence Microscopy Experiments. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2016; 22:538-539. [PMID: 28386206 PMCID: PMC5380232 DOI: 10.1017/s1431927616003548] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Affiliation(s)
- Allen Goodman
- Imaging Platform, Broad Institute of Harvard and MIT, Cambridge USA
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of Harvard and MIT, Cambridge USA
| |
Collapse
|
163
|
Quan Y, Xiong L, Chen J, Zhang HY. Genetics-directed drug discovery for combating Mycobacterium tuberculosis infection. J Biomol Struct Dyn 2016; 35:616-621. [DOI: 10.1080/07391102.2016.1157037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Yuan Quan
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Le Xiong
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Jing Chen
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| |
Collapse
|
164
|
Schaaf K, Hayley V, Speer A, Wolschendorf F, Niederweis M, Kutsch O, Sun J. A Macrophage Infection Model to Predict Drug Efficacy Against Mycobacterium Tuberculosis. Assay Drug Dev Technol 2016; 14:345-54. [PMID: 27327048 DOI: 10.1089/adt.2016.717] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
In the last 40 years, only a single new antituberculosis drug was FDA approved. New tools that improve the drug development process will be essential to accelerate the development of next-generation antituberculosis drugs. The drug development process seems to be hampered by the inefficient transition of initially promising hits to candidate compounds that are effective in vivo. In this study, we introduce an inexpensive, rapid, and BSL-2 compatible infection model using macrophage-passaged Mycobacterium tuberculosis (Mtb) that forms densely packed Mtb/macrophage aggregate structures suitable for drug efficacy testing. Susceptibility to antituberculosis drugs determined with this Mtb/macrophage aggregate model differed from commonly used in vitro broth-grown single-cell Mtb cultures. Importantly, altered drug susceptibility correlated well with the reported ability of the respective drugs to generate high tissue and cerebrospinal fluid concentrations relative to their serum concentrations, which seems to be the best predictors of in vivo efficacy. Production of these Mtb/macrophage aggregates could be easily scaled up to support throughput efforts. Overall, its simplicity and scalability should make this Mtb/macrophage aggregate model a valuable addition to the currently available Mtb drug discovery tools.
Collapse
Affiliation(s)
- Kaitlyn Schaaf
- 1 Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Virginia Hayley
- 1 Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Alexander Speer
- 2 Department of Microbiology, University of Alabama at Birmingham , Birmingham, Alabama.,3 Department of Medical Microbiology and Infection Control, VU University Medical Center , Amsterdam, Netherlands
| | - Frank Wolschendorf
- 1 Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Michael Niederweis
- 2 Department of Microbiology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Olaf Kutsch
- 1 Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Jim Sun
- 2 Department of Microbiology, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
165
|
Ma J, Jung BG, Yi N, Samten B. Early Secreted Antigenic Target of 6 kDa ofMycobacterium tuberculosisStimulates Macrophage Chemoattractant Protein-1 Production by Macrophages and Its Regulation by p38 Mitogen-Activated Protein Kinases and Interleukin-4. Scand J Immunol 2016; 84:39-48. [DOI: 10.1111/sji.12447] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 05/04/2016] [Indexed: 11/29/2022]
Affiliation(s)
- J. Ma
- Department of Pulmonary Immunology; University of Texas Health Science Center at Tyler; Tyler TX USA
| | - B-G. Jung
- Department of Pulmonary Immunology; University of Texas Health Science Center at Tyler; Tyler TX USA
| | - N. Yi
- Department of Pulmonary Immunology; University of Texas Health Science Center at Tyler; Tyler TX USA
| | - B. Samten
- Department of Pulmonary Immunology; University of Texas Health Science Center at Tyler; Tyler TX USA
| |
Collapse
|
166
|
Ang MLT, Pethe K. Contribution of high-content imaging technologies to the development of anti-infective drugs. Cytometry A 2016; 89:755-60. [PMID: 27272127 PMCID: PMC5089693 DOI: 10.1002/cyto.a.22885] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/17/2016] [Accepted: 05/06/2016] [Indexed: 12/18/2022]
Abstract
Originally developed to study fundamental aspects of cellular biology, high‐content imaging (HCI) was rapidly adapted to study host–pathogen interactions at the cellular level and adopted as a technology of choice to unravel disease biology. HCI platforms allow for the visualization and quantification of discrete phenotypes that cannot be captured using classical screening approaches. A key advantage of high‐content screening technologies lies in the possibility to develop and interrogate physiologically significant, predictive ex vivo disease models that reproduce complex conditions relevant for infection. Here we review and discuss recent advances in HCI technologies and chemical biology approaches that are contributing to an increased understanding of the intricate host–pathogen interrelationship on the cellular level, and which will foster the development of novel therapeutic approaches for the treatment of human bacterial and protozoan infections. © 2016 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of ISAC
Collapse
Affiliation(s)
- Michelle Lay Teng Ang
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, Singapore
| | - Kevin Pethe
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
167
|
Yang H, Liu H, Chen H, Mo H, Chen J, Huang X, Zheng R, Liu Z, Feng Y, Liu F, Ge B. G protein-coupled receptor160 regulates mycobacteria entry into macrophages by activating ERK. Cell Signal 2016; 28:1145-1151. [PMID: 27259691 DOI: 10.1016/j.cellsig.2016.05.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 05/22/2016] [Accepted: 05/29/2016] [Indexed: 11/19/2022]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, invades and replicates within susceptible hosts by disturbing host antimicrobial mechanisms. Although G protein-coupled receptors (GPCRs) are involved in most physiological and pathological activities of mammalian cells, the roles of GPCRs in Mtb invasion into host cell remain elusive. Here, we report that GPR160 expression is elevated at both mRNA and protein level in macrophages in response to BCG infection. Both the PiggyBac (PB) transposon-mediated mutation of gpr160 gene in mouse primary macrophages and siRNA-mediated knockdown of GPR160 in the human macrophage cell line THP-1 markedly reduced the entry of green fluorescent protein (GFP) expressing BCG (BCG-GFP), also operative in vivo. BCG infection-induced phosphorylation of ERK1/2 was significantly reduced in gpr160 mutated (gpr160(-/-)) macrophages relative to levels observed in wild type macrophages, while inhibition of ERK by specific inhibitor or knockdown ERK1/2 by specific siRNA markedly reduced entry of BCG. Finally, lower bacteria burdens and attenuated pathological impairments were observed in the lungs of BCG-infected gpr160(-/-) mice. Furthermore, gpr160(-/-) macrophages also exhibits reduced uptake of Escherichia coli and Francisella tularensis. Taken together, these findings suggest an important role of GPR160 in regulating the entry of BCG into macrophages by targeting the ERK signaling pathway. As GPCRs have proven to be successful drug targets in pharmaceutical industry, it's tempting to speculate that compounds targeting GPR160, a G protein-coupled receptor, could intervene in Mtb infection.
Collapse
Affiliation(s)
- Hua Yang
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine,507 Zhengmin Road, Shanghai 200433, PR China
| | - Haipeng Liu
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine,507 Zhengmin Road, Shanghai 200433, PR China
| | - Hao Chen
- Department of Microbiology and Immunology, Tongji University School of Medicine,1239 Siping Road, Shanghai 200049, PR China
| | - Haiping Mo
- Department of Microbiology and Immunology, Tongji University School of Medicine,1239 Siping Road, Shanghai 200049, PR China
| | - Jianxia Chen
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine,507 Zhengmin Road, Shanghai 200433, PR China
| | - Xiaocheng Huang
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine,507 Zhengmin Road, Shanghai 200433, PR China
| | - Ruijuan Zheng
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine,507 Zhengmin Road, Shanghai 200433, PR China
| | - Zhonghua Liu
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine,507 Zhengmin Road, Shanghai 200433, PR China
| | - Yonghong Feng
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine,507 Zhengmin Road, Shanghai 200433, PR China
| | - Feng Liu
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine,507 Zhengmin Road, Shanghai 200433, PR China
| | - Baoxue Ge
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine,507 Zhengmin Road, Shanghai 200433, PR China; Department of Microbiology and Immunology, Tongji University School of Medicine,1239 Siping Road, Shanghai 200049, PR China.
| |
Collapse
|
168
|
Samantaray S, Correia JN, Garelnabi M, Voelz K, May RC, Hall RA. Novel cell-based in vitro screen to identify small-molecule inhibitors against intracellular replication of Cryptococcus neoformans in macrophages. Int J Antimicrob Agents 2016; 48:69-77. [PMID: 27289450 PMCID: PMC4942879 DOI: 10.1016/j.ijantimicag.2016.04.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/07/2016] [Accepted: 04/16/2016] [Indexed: 01/08/2023]
Abstract
The fungal pathogen Cryptococcus neoformans poses a major threat to immunocompromised patients and is a leading killer of human immunodeficiency virus (HIV)-infected patients worldwide. Cryptococci are known to manipulate host macrophages and can either remain latent or proliferate intracellularly within the host phagocyte, a favourable niche that also renders them relatively insensitive to antifungal agents. Here we report an attempt to address this limitation by using a fluorescence-based drug screening method to identify potential inhibitors of intracellular proliferation of C. neoformans. The Prestwick Chemical Library(®) of FDA-approved small molecules was screened for compounds that limit the intracellular replication of a fluorescently-tagged C. neoformans reference strain (H99-GFP) in macrophages. Preliminary screening revealed 19 of 1200 compounds that could significantly reduce intracellular growth of the pathogen. Secondary screening and host cell cytotoxicity assays highlighted fendiline hydrochloride as a potential drug candidate for the development of future anticryptococcal therapies. Live cell imaging demonstrated that this Ca(2+) channel blocker strongly enhanced phagosome maturation in macrophages leading to improved fungal killing and reduced intracellular replication. Whilst the relatively high dose of fendiline hydrochloride required renders it unfit for clinical deployment against cryptococcosis, this study highlights a novel approach for identifying new lead compounds and unravels a pharmacologically promising scaffold towards the development of novel antifungal therapies for this neglected disease.
Collapse
Affiliation(s)
- Sweta Samantaray
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Joao N Correia
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Mariam Garelnabi
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Kerstin Voelz
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Robin C May
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK.
| | - Rebecca A Hall
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
169
|
Dean SN, van Hoek ML. Screen of FDA-approved drug library identifies maprotiline, an antibiofilm and antivirulence compound with QseC sensor-kinase dependent activity in Francisella novicida. Virulence 2016; 6:487-503. [PMID: 26155740 DOI: 10.1080/21505594.2015.1046029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Development of new therapeutics against Select Agents such as Francisella is critical preparation in the event of bioterrorism. Testing FDA-approved drugs for this purpose may yield new activities unrelated to their intended purpose and may hasten the discovery of new therapeutics. A library of 420 FDA-approved drugs was screened for antibiofilm activity against a model organism for human tularemia, Francisella (F.) novicida, excluding drugs that significantly inhibited growth. The initial screen was based on the 2-component system (TCS) dependent biofilm effect, thus, the QseC dependence of maprotiline anti-biofilm action was demonstrated. By comparing their FDA-approved uses, chemical structures, and other properties of active drugs, toremifene and polycyclic antidepressants maprotiline and chlorpromazine were identified as being highly active against F. novicida biofilm formation. Further down-selection excluded toremifene for its membrane active activity and chlorpromazine for its high antimicrobial activity. The mode of action of maprotiline against F. novicida was sought. It was demonstrated that maprotiline was able to significantly down-regulate the expression of the virulence factor IglC, encoded on the Francisella Pathogenicity Island (FPI), suggesting that maprotiline is exerting an effect on bacterial virulence. Further studies showed that maprotiline significantly rescued F. novicida infected wax worm larvae. In vivo studies demonstrated that maprotiline treatment could prolong time to disease onset and survival in F. novicida infected mice. These results suggest that an FDA-approved drug such as maprotiline has the potential to combat Francisella infection as an antivirulence agent, and may have utility in combination with antibiotics.
Collapse
Affiliation(s)
- Scott N Dean
- a National Center for Biodefense and Infectious Diseases ; Manassas , VA , USA
| | | |
Collapse
|
170
|
Abstract
Tuberculosis (TB) is still a major global health problem. A third of the world’s population is infected with Mycobacterium tuberculosis. Only ~10% of infected individuals develop TB but there are 9 million TB cases with 1.5 million deaths annually. The standard prophylactic treatment regimens for latent TB infection take 3–9 months, and new cases of TB require at least 6 months of treatment with multiple drugs. The management of latent TB infection and TB has become more challenging because of the spread of multidrug-resistant and extremely drug-resistant TB. Intensified efforts to find new TB drugs and immunotherapies are needed. Immunotherapies could modulate the immune system in patients with latent TB infection or active disease, enabling better control of M. tuberculosis replication. This review describes several types of potential immunotherapies with a focus on those which have been tested in humans.
Collapse
Affiliation(s)
- Getahun Abate
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology
| | - Daniel F Hoft
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology; Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, USA
| |
Collapse
|
171
|
Bhattacharya B, Chatterjee S, Devine WG, Kobzik L, Beeler AB, Porco JA, Kramnik I. Fine-tuning of macrophage activation using synthetic rocaglate derivatives. Sci Rep 2016; 6:24409. [PMID: 27086720 PMCID: PMC4834551 DOI: 10.1038/srep24409] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 03/29/2016] [Indexed: 12/25/2022] Open
Abstract
Drug-resistant bacteria represent a significant global threat. Given the dearth of new antibiotics, host-directed therapies (HDTs) are especially desirable. As IFN-gamma (IFNγ) plays a central role in host resistance to intracellular bacteria, including Mycobacterium tuberculosis, we searched for small molecules to augment the IFNγ response in macrophages. Using an interferon-inducible nuclear protein Ipr1 as a biomarker of macrophage activation, we performed a high-throughput screen and identified molecules that synergized with low concentration of IFNγ. Several active compounds belonged to the flavagline (rocaglate) family. In primary macrophages a subset of rocaglates 1) synergized with low concentrations of IFNγ in stimulating expression of a subset of IFN-inducible genes, including a key regulator of the IFNγ network, Irf1; 2) suppressed the expression of inducible nitric oxide synthase and type I IFN and 3) induced autophagy. These compounds may represent a basis for macrophage-directed therapies that fine-tune macrophage effector functions to combat intracellular pathogens and reduce inflammatory tissue damage. These therapies would be especially relevant to fighting drug-resistant pathogens, where improving host immunity may prove to be the ultimate resource.
Collapse
Affiliation(s)
- Bidisha Bhattacharya
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, 02118, USA
| | - Sujoy Chatterjee
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, 02118, USA
| | - William G Devine
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, 02215, USA
| | - Lester Kobzik
- Department of Environmental Health, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Aaron B Beeler
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, 02215, USA
| | - John A Porco
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, 02215, USA
| | - Igor Kramnik
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, 02118, USA
| |
Collapse
|
172
|
Ouimet M, Koster S, Sakowski E, Ramkhelawon B, van Solingen C, Oldebeken S, Karunakaran D, Portal-Celhay C, Sheedy FJ, Ray TD, Cecchini K, Zamore PD, Rayner KJ, Marcel YL, Philips JA, Moore KJ. Mycobacterium tuberculosis induces the miR-33 locus to reprogram autophagy and host lipid metabolism. Nat Immunol 2016; 17:677-86. [PMID: 27089382 PMCID: PMC4873392 DOI: 10.1038/ni.3434] [Citation(s) in RCA: 279] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 03/11/2016] [Indexed: 12/16/2022]
Abstract
Mycobacterium tuberculosis (Mtb) survives in macrophages by evading delivery to the lysosome and promoting the accumulation of lipid bodies, which serve as a bacterial source of nutrients. We found that by inducing the microRNA (miRNA) miR-33 and its passenger strand miR-33*, Mtb inhibited integrated pathways involved in autophagy, lysosomal function and fatty acid oxidation to support bacterial replication. Silencing of miR-33 and miR-33* by genetic or pharmacological means promoted autophagy flux through derepression of key autophagy effectors (such as ATG5, ATG12, LC3B and LAMP1) and AMPK-dependent activation of the transcription factors FOXO3 and TFEB, which enhanced lipid catabolism and Mtb xenophagy. These data define a mammalian miRNA circuit used by Mtb to coordinately inhibit autophagy and reprogram host lipid metabolism to enable intracellular survival and persistence in the host.
Collapse
Affiliation(s)
- Mireille Ouimet
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Stefan Koster
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Erik Sakowski
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Bhama Ramkhelawon
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Coen van Solingen
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Scott Oldebeken
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Denuja Karunakaran
- University of Ottawa Heart Institute and Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Cynthia Portal-Celhay
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Frederick J Sheedy
- Department of Clinical Medicine, School of Medicine, Trinity College, Dublin, Ireland
| | - Tathagat Dutta Ray
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Katharine Cecchini
- RNA Therapeutics Institute, Howard Hughes Medical Institute, and Department of Biochemistry &Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Philip D Zamore
- RNA Therapeutics Institute, Howard Hughes Medical Institute, and Department of Biochemistry &Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Katey J Rayner
- University of Ottawa Heart Institute and Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Yves L Marcel
- University of Ottawa Heart Institute and Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Jennifer A Philips
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Kathryn J Moore
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| |
Collapse
|
173
|
Targeting Drug-Sensitive and -Resistant Strains of Mycobacterium tuberculosis by Inhibition of Src Family Kinases Lowers Disease Burden and Pathology. mSphere 2016; 1:mSphere00043-15. [PMID: 27303736 PMCID: PMC4894694 DOI: 10.1128/msphere.00043-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 03/14/2016] [Indexed: 11/30/2022] Open
Abstract
The existing treatment regimen for tuberculosis (TB) suffers from deficiencies like high doses of antibiotics, long treatment duration, and inability to kill persistent populations in an efficient manner. Together, these contribute to the emergence of drug-resistant tuberculosis. Recently, several host factors were identified which help intracellular survival of Mycobacterium tuberculosis within the macrophage. These factors serve as attractive targets for developing alternate therapeutic strategies against M. tuberculosis. This strategy promises to be effective against drug-resistant strains. The approach also has potential to considerably lower the risk of emergence of new drug-resistant strains. We explored tyrosine kinase Src as a host factor exploited by virulent M. tuberculosis for intracellular survival. We show that Src inhibition can effectively control tuberculosis in infected guinea pigs. Moreover, Src inhibition ameliorated TB-associated pathology in guinea pigs. Thus, Src inhibitors have strong potential to be developed as possible anti-TB drugs. In view of emerging drug resistance among bacterial pathogens, including Mycobacterium tuberculosis, the development of novel therapeutic strategies is increasingly being sought. A recent paradigm in antituberculosis (anti-TB) drug development is to target the host molecules that are crucial for intracellular survival of the pathogen. We previously showed the importance of Src tyrosine kinases in mycobacterial pathogenesis. Here, we report that inhibition of Src significantly reduced survival of H37Rv as well as multidrug-resistant (MDR) and extremely drug-resistant (XDR) strains of M. tuberculosis in THP-1 macrophages. Src inhibition was also effective in controlling M. tuberculosis infection in guinea pigs. In guinea pigs, reduced M. tuberculosis burden due to Src inhibition also led to a marked decline in the disease pathology. In agreement with the theoretical framework of host-directed approaches against the pathogen, Src inhibition was equally effective against an XDR strain in controlling infection in guinea pigs. We propose that Src inhibitors could be developed into effective host-directed anti-TB drugs, which could be indiscriminately used against both drug-sensitive and drug-resistant strains of M. tuberculosis. IMPORTANCE The existing treatment regimen for tuberculosis (TB) suffers from deficiencies like high doses of antibiotics, long treatment duration, and inability to kill persistent populations in an efficient manner. Together, these contribute to the emergence of drug-resistant tuberculosis. Recently, several host factors were identified which help intracellular survival of Mycobacterium tuberculosis within the macrophage. These factors serve as attractive targets for developing alternate therapeutic strategies against M. tuberculosis. This strategy promises to be effective against drug-resistant strains. The approach also has potential to considerably lower the risk of emergence of new drug-resistant strains. We explored tyrosine kinase Src as a host factor exploited by virulent M. tuberculosis for intracellular survival. We show that Src inhibition can effectively control tuberculosis in infected guinea pigs. Moreover, Src inhibition ameliorated TB-associated pathology in guinea pigs. Thus, Src inhibitors have strong potential to be developed as possible anti-TB drugs.
Collapse
|
174
|
Muciño G, Castro-Obregón S, Hernandez-Pando R, Del Rio G. Autophagy as a target for therapeutic uses of multifunctional peptides. IUBMB Life 2016; 68:259-67. [PMID: 26968336 DOI: 10.1002/iub.1483] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/17/2016] [Indexed: 12/21/2022]
Abstract
The emergence of complex diseases is promoting a change from one-target to multitarget drugs and peptides are ideal molecules to fulfill this polypharmacologic role. Here we review current status in the design of polypharmacological peptides aimed to treat complex diseases, focusing on tuberculosis. In this sense, combining multiple activities in single molecules is a two-sided sword, as both positive and negative side effects might arise. These polypharmacologic compounds may be directed to regulate autophagy, a catabolic process that enables cells to eliminate intracellular microbes (xenophagy), such as Mycobacterium tuberculosis (MBT). Here we review some strategies to control MBT infection and propose that a peptide combining both antimicrobial and pro-autophagic activities would have a greater potential to limit MBT infection. This endeavor may complement the knowledge gained in understanding the mechanism of action of antibiotics and may lead to the design of better polypharmacological peptides to treat complex diseases such as tuberculosis.
Collapse
Affiliation(s)
- Gabriel Muciño
- Department of Neurodevelopment and Physiology, Instituto De Fisiología Celular, UNAM, México, D.F, México
| | - Susana Castro-Obregón
- Department of Neurodevelopment and Physiology, Instituto De Fisiología Celular, UNAM, México, D.F, México
| | - Rogelio Hernandez-Pando
- Experimental Pathology Section, Department of Pathology, National Institute of Medical Sciences and Nutrition "Salvador Zubirán,", Mexico
| | - Gabriel Del Rio
- Department of Biochemistry and Structural Biology, Instituto De Fisiología Celular, UNAM, México, D.F, México
| |
Collapse
|
175
|
Kumar R, Sahu SK, Kumar M, Jana K, Gupta P, Gupta UD, Kundu M, Basu J. MicroRNA 17-5p regulates autophagy in Mycobacterium tuberculosis-infected macrophages by targeting Mcl-1 and STAT3. Cell Microbiol 2015; 18:679-91. [PMID: 26513648 DOI: 10.1111/cmi.12540] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 10/23/2015] [Accepted: 10/23/2015] [Indexed: 12/30/2022]
Abstract
Autophagy plays a crucial role in the control of bacterial burden during Mycobacterium tuberculosis infection. MicroRNAs (miRNAs) are small non-coding RNAs that regulate immune signalling and inflammation in response to challenge by pathogens. Appreciating the potential of host-directed therapies designed to control autophagy during mycobacterial infection, we focused on the role of miRNAs in regulating M. tuberculosis-induced autophagy in macrophages. Here, we demonstrate that M. tuberculosis infection leads to downregulation of miR-17 and concomitant upregulation of its targets Mcl-1 and STAT3, a transcriptional activator of Mcl-1. Forced expression of miR-17 reduces expression of Mcl-1 and STAT3 and also the interaction between Mcl-1 and Beclin-1. This is directly linked to enhanced autophagy, because Mcl-1 overexpression attenuates the effects of miR-17. At the same time, transfection with a kinase-inactive mutant of protein kinase C δ (PKCδ) (an activator of STAT3) augments M. tuberculosis-induced autophagy, and miR-17 overexpression diminishes phosphorylation of PKCδ, suggesting that an miR-17/PKC δ/STAT3 axis regulates autophagy during M. tuberculosis infection.
Collapse
Affiliation(s)
- Ranjeet Kumar
- Department of Chemistry, Bose Institute, Kolkata, 700009, India
| | | | - Manish Kumar
- Department of Chemistry, Bose Institute, Kolkata, 700009, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata, 700054, India
| | - Pushpa Gupta
- National Jalma Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 282006, India
| | - Umesh D Gupta
- National Jalma Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 282006, India
| | | | - Joyoti Basu
- Department of Chemistry, Bose Institute, Kolkata, 700009, India
| |
Collapse
|
176
|
Doyle SK, Pop MS, Evans HL, Koehler AN. Advances in discovering small molecules to probe protein function in a systems context. Curr Opin Chem Biol 2015; 30:28-36. [PMID: 26615565 DOI: 10.1016/j.cbpa.2015.10.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/30/2015] [Accepted: 10/30/2015] [Indexed: 12/15/2022]
Abstract
High throughput screening (HTS) has historically been used for drug discovery almost exclusively by the pharmaceutical industry. Due to a significant decrease in costs associated with establishing a high throughput facility and an exponential interest in discovering probes of development and disease associated biomolecules, HTS core facilities have become an integral part of most academic and non-profit research institutions over the past decade. This major shift has led to the development of new HTS methodologies extending beyond the capabilities and target classes used in classical drug discovery approaches such as traditional enzymatic activity-based screens. In this brief review we describe some of the most interesting developments in HTS technologies and methods for chemical probe discovery.
Collapse
Affiliation(s)
- Shelby K Doyle
- David H. Koch Institute for Integrative Cancer Research, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marius S Pop
- David H. Koch Institute for Integrative Cancer Research, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Helen L Evans
- David H. Koch Institute for Integrative Cancer Research, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Angela N Koehler
- David H. Koch Institute for Integrative Cancer Research, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
177
|
Hawn TR, Shah JA, Kalman D. New tricks for old dogs: countering antibiotic resistance in tuberculosis with host-directed therapeutics. Immunol Rev 2015; 264:344-62. [PMID: 25703571 DOI: 10.1111/imr.12255] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite the availability of Mycobacterium tuberculosis (Mtb) drugs for over 50 years, tuberculosis (TB) remains at pandemic levels. New drugs are urgently needed for resistant strains, shortening duration of treatment, and targeting different stages of the disease, especially for treatment during human immunodeficiency virus co-infection. One solution to the conundrum that antibiotics kill the bacillus yet select for resistance is to target the host rather than the pathogen. Here, we discuss recent progress in so-called 'host-directed therapeutics' (HDTs), focusing on two general mechanistic strategies: (i) HDTs that disrupt Mtb pathogenesis in macrophages and (ii) immunomodulatory HDTs that facilitate protective immune responses that kill Mtb or reduce deleterious responses that exacerbate disease. HDTs hold significant promise as adjunctive therapies in that they are less likely to engender resistance, will likely have efficacy against antibiotic-resistant strains, and may have activity against non-replicating Mtb. However, TB is a complex and variegated disease, and human populations exhibit significant diversity in their immune responses to it, which presents a complicated landscape for HDTs to navigate. Nevertheless, we suggest that a detailed mechanistic understanding of drug action, together with careful selection of disease stage targets and dosing strategies may overcome such limitations and allow the development of HDTs as effective adjunctive treatment options for TB.
Collapse
Affiliation(s)
- Thomas R Hawn
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
178
|
Baer CE, Rubin EJ, Sassetti CM. New insights into TB physiology suggest untapped therapeutic opportunities. Immunol Rev 2015; 264:327-43. [PMID: 25703570 DOI: 10.1111/imr.12267] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The current regimens used to treat tuberculosis are largely comprised of serendipitously discovered drugs that are combined based on clinical experience. Despite curing millions, these drug regimens are limited by the long course of therapy, the emergence of resistance, and the persistent tissue damage that remains after treatment. The last two decades have produced only a single new drug but have represented a renaissance in our understanding of the physiology of tuberculosis infection. The advent of mycobacterial genetics, sophisticated immunological methods, and imaging technologies have transformed our understanding of bacterial physiology as well as the contribution of the host response to disease outcome. Specific alterations in bacterial metabolism, heterogeneity in bacterial state, and drug penetration all limit the effectiveness of antimicrobial therapy. This review summarizes these new biological insights and discusses strategies to exploit them for the rational development of more effective therapeutics. Three general strategies are discussed. First, our emerging insight into bacterial physiology suggests new pathways that might be targeted to accelerate therapy. Second, we explore whether the concept of genetic synergy can be used to design effective combination therapies. Finally, we outline possible approaches to modulate the host response to accentuate antibiotic efficacy. These biology-driven strategies promise to produce more effective therapies.
Collapse
Affiliation(s)
- Christina E Baer
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | | | |
Collapse
|
179
|
Development of an Intracellular Screen for New Compounds Able To Inhibit Mycobacterium tuberculosis Growth in Human Macrophages. Antimicrob Agents Chemother 2015; 60:640-5. [PMID: 26503663 DOI: 10.1128/aac.01920-15] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/23/2015] [Indexed: 12/19/2022] Open
Abstract
Here we describe the development and validation of an intracellular high-throughput screening assay for finding new antituberculosis compounds active in human macrophages. The assay consists of a luciferase-based primary identification assay, followed by a green fluorescent protein-based secondary profiling assay. Standard tuberculosis drugs and 158 previously recognized active antimycobacterial compounds were used to evaluate assay robustness. Data show that the assay developed is a short and valuable tool for the discovery of new antimycobacterial compounds.
Collapse
|
180
|
Genome Expression Profiling-Based Identification and Administration Efficacy of Host-Directed Antimicrobial Drugs against Respiratory Infection by Nontypeable Haemophilus influenzae. Antimicrob Agents Chemother 2015; 59:7581-92. [PMID: 26416856 DOI: 10.1128/aac.01278-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/20/2015] [Indexed: 12/15/2022] Open
Abstract
Therapies that are safe, effective, and not vulnerable to developing resistance are highly desirable to counteract bacterial infections. Host-directed therapeutics is an antimicrobial approach alternative to conventional antibiotics based on perturbing host pathways subverted by pathogens during their life cycle by using host-directed drugs. In this study, we identified and evaluated the efficacy of a panel of host-directed drugs against respiratory infection by nontypeable Haemophilus influenzae (NTHi). NTHi is an opportunistic pathogen that is an important cause of exacerbation of chronic obstructive pulmonary disease (COPD). We screened for host genes differentially expressed upon infection by the clinical isolate NTHi375 by analyzing cell whole-genome expression profiling and identified a repertoire of host target candidates that were pharmacologically modulated. Based on the proposed relationship between NTHi intracellular location and persistence, we hypothesized that drugs perturbing host pathways used by NTHi to enter epithelial cells could have antimicrobial potential against NTHi infection. Interfering drugs were tested for their effects on bacterial and cellular viability, on NTHi-epithelial cell interplay, and on mouse pulmonary infection. Glucocorticoids and statins lacked in vitro and/or in vivo efficacy. Conversely, the sirtuin-1 activator resveratrol showed a bactericidal effect against NTHi, and the PDE4 inhibitor rolipram showed therapeutic efficacy by lowering NTHi375 counts intracellularly and in the lungs of infected mice. PDE4 inhibition is currently prescribed in COPD, and resveratrol is an attractive geroprotector for COPD treatment. Together, these results expand our knowledge of NTHi-triggered host subversion and frame the antimicrobial potential of rolipram and resveratrol against NTHi respiratory infection.
Collapse
|
181
|
Protection and pathology in TB: learning from the zebrafish model. Semin Immunopathol 2015; 38:261-73. [PMID: 26324465 PMCID: PMC4779130 DOI: 10.1007/s00281-015-0522-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/11/2015] [Indexed: 12/14/2022]
Abstract
Zebrafish has earned its place among animal models of tuberculosis. Its natural pathogen, Mycobacterium marinum, shares major virulence factors with the human pathogen Mycobacterium tuberculosis. In adult zebrafish, which possess recombination-activated adaptive immunity, it can cause acute infection or a chronic progressive disease with containment of mycobacteria in well-structured, caseating granulomas. In addition, a low-dose model that closely mimics human latent infection has recently been developed. These models are used alongside infection of optically transparent zebrafish embryos and larvae that rely on innate immunity and permit non-invasive visualization of the early stages of developing granulomas that are inaccessible in other animal models. By microinjecting mycobacteria intravenously or into different tissues, systemic and localized infections can be induced, each useful for studying particular aspects of early pathogenesis, such as phagocyte recruitment, granuloma expansion and maintenance, vascularization of granulomas, and the phagocyte-mediated dissemination of mycobacteria. This has contributed to new insights into the mycobacteria-driven mechanisms that promote granuloma formation, the double-edged role of inflammation, the mechanisms of macrophage cell death that favor disease progression, and the host-protective role of autophagy. As a result, zebrafish models are now increasingly used to explore strategies for adjunctive therapy of tuberculosis with host-directed drugs.
Collapse
|
182
|
Selective enrichment of mycobacterial proteins from infected host macrophages. Sci Rep 2015; 5:13430. [PMID: 26303024 PMCID: PMC4548221 DOI: 10.1038/srep13430] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/27/2015] [Indexed: 12/20/2022] Open
Abstract
Upon infection, Mycobacterium tuberculosis (Mtb) deploys specialized secretion machinery to deliver virulent proteins with the capacity to modulate a variety of host-cellular pathways. Studies on the identification of intra-macrophage Mtb proteins, however, are constricted by an inability to selectively enrich these virulent effectors against overwhelming protein content of the host. Here, we introduce an Mtb-selective protein labeling method based on genetic incorporation of azidonorleucine (Anl) through the expression of a mutant methionyl-tRNA synthetase. Exclusive incorporation of Anl, into native Mtb proteins, provided a click handle to pull out low abundant secretory proteins from the lysates of infected cells. Further, temporal secretome profiling, upon infection with strains of varying degree of virulence, revealed the proficiency of virulent Mtb to secrete chaperones. This ability contributed at least partially to the mycobacterial virulence-specific suppression of ER stress in the host macrophage, representing an important facet of mycobacterial virulence. The Anl labeling approach should facilitate new exciting opportunities for imaging and proteomic investigations of differently virulent Mtb isolates to understand determinants of pathogenicity.
Collapse
|
183
|
Zuniga ES, Early J, Parish T. The future for early-stage tuberculosis drug discovery. Future Microbiol 2015; 10:217-29. [PMID: 25689534 DOI: 10.2217/fmb.14.125] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
There is an urgent need for new and better drugs to treat tuberculosis due to lengthy and complex treatment regimens and a rising problem of drug resistance. Drug discovery efforts have increased over the past few years, with a larger focus on modern high-throughput screening technologies. A combination of target-based approaches, with the traditional empirical means of drug identification, has been complemented by the use of target-based phenotypic screens only recently made possibly with newer genetic tools. Using these approaches, a number of promising compound series have been discovered. However, significant problems remain in developing these into drugs. This review highlights recent advances in TB drug discovery, including an overview of screening campaigns, lessons learned and future directions.
Collapse
Affiliation(s)
- Edison S Zuniga
- TB Discovery Research, Infectious Disease Research Institute, Seattle, WA 98102, USA
| | | | | |
Collapse
|
184
|
Ang MLT, Murima P, Pethe K. Next-generation antimicrobials: from chemical biology to first-in-class drugs. Arch Pharm Res 2015; 38:1702-17. [PMID: 26259630 PMCID: PMC4567591 DOI: 10.1007/s12272-015-0645-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/29/2015] [Indexed: 01/11/2023]
Abstract
The global emergence of multi-drug resistant bacteria invokes an urgent and imperative necessity for the identification of novel antimicrobials. The general lack of success in progressing novel chemical entities from target-based drug screens have prompted calls for radical and innovative approaches for drug discovery. Recent developments in chemical biology and target deconvolution strategies have revived interests in the utilization of whole-cell phenotypic screens and resulted in several success stories for the discovery and development novel drug candidates and target pathways. In this review, we present and discuss recent chemical biology approaches focusing on the discovery of novel targets and new lead molecules for the treatment of human bacterial and protozoan infections.
Collapse
Affiliation(s)
- Michelle Lay Teng Ang
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, 30 Biopolis Street, #B2-15a, Singapore, 138671, Singapore.
| | - Paul Murima
- Global Health Institute, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Kevin Pethe
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, 30 Biopolis Street, #B2-15a, Singapore, 138671, Singapore.
| |
Collapse
|
185
|
Zumla A, Maeurer M, Chakaya J, Hoelscher M, Ntoumi F, Rustomjee R, Vilaplana C, Yeboah-Manu D, Rasolof V, Munderi P, Singh N, Aklillu E, Padayatchi N, Macete E, Kapata N, Mulenga M, Kibiki G, Mfinanga S, Nyirenda T, Maboko L, Garcia-Basteiro A, Rakotosamimanana N, Bates M, Mwaba P, Reither K, Gagneux S, Edwards S, Mfinanga E, Abdulla S, Cardona PJ, Russell JBW, Gant V, Noursadeghi M, Elkington P, Bonnet M, Menendez C, Dieye TN, Diarra B, Maiga A, Aseffa A, Parida S, Wejse C, Petersen E, Kaleebu P, Oliver M, Craig G, Corrah T, Tientcheu L, Antonio M, Rao M, McHugh TD, Sheikh A, Ippolito G, Ramjee G, Kaufmann SHE, Churchyard G, Steyn A, Grobusch M, Sanne I, Martinson N, Madansein R, Wilkinson RJ, Mayosi B, Schito M, Wallis RS. Towards host-directed therapies for tuberculosis. Nat Rev Drug Discov 2015; 14:511-2. [PMID: 26184493 DOI: 10.1038/nrd4696] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The treatment of tuberculosis is based on combinations of drugs that directly target Mycobacterium tuberculosis. A new global initiative is now focusing on a complementary approach of developing adjunct host-directed therapies.
Collapse
Affiliation(s)
- Alimuddin Zumla
- 1] Division of Infection and Immunity at the University College London (UCL), London, UK and National Institute of Health Research's Biomedical Research Centre UCL Hospitals National Health Service Foundation Trust, Royal Free Campus, 2nd Floor, Rowland Hill Street, London NW3 2PF, UK. [2] Department of Microbiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Markus Maeurer
- Therapeutic Immunology, Department of Laboratory Medicine, Karolinska Institutet and Center for Allogeneic Stem Cell Transplantation, Karolinska Hospital, Hälsovägen, SE-14186 Stockholm, Sweden
| | | | | | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, Medical Centre of the University of Munich (LMU), and DZIF German Centre for Infection Research, Munich, Germany
| | - Francine Ntoumi
- Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo and Institute for Tropical Medicine, University of Tübingen, Germany
| | | | - Cristina Vilaplana
- Unitat de Tuberculosi Experimental Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i PujolEdifici Laboratoris de Recerca Can Ruti Campus, Barcelona, Spain
| | - Dorothy Yeboah-Manu
- Bacteriology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Voahangy Rasolof
- Mycobacteria Unit, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | - Paula Munderi
- Medical Research Council/Uganda Virus Research Institute, Research Unit on AIDS, Entebbe, Uganda
| | - Nalini Singh
- Inkosi Albert Luthuli Central Hospital and King DinuZulu Hospital, Durban, South Africa
| | - Eleni Aklillu
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nesri Padayatchi
- Centre for AIDS Prevention Research in South Africa (CAPRISA), SAMRC CAPRISA HIV -TB pathogenesis and treatment research unit, Durban, South Africa
| | - Eusebio Macete
- Centro de Investigação em Saude de Manhiça, Maputo, Mozambique
| | - Nathan Kapata
- UNZA-UCLMS Research and Training Project, University Teaching Hospital, Lusaka, Zambia
| | | | - Gibson Kibiki
- Kilimanjaro Clinical Research Institute (KCRI), Kilimanjaro, Tanzania
| | - Sayoki Mfinanga
- Muhimbili Medical Research Centre National Institute for Medical Research (NIMR), Dar es Salaam, Tanzania
| | - Thomas Nyirenda
- European Developing Countries Clinical Trials Partnership (EDCTP), Cape Town, South Africa; not officially a partner of this consortium but participated in the HDT-NET Cape Town launch
| | | | | | | | - Matthew Bates
- UNZA-UCLMS Research and Training Project, University Teaching Hospital, Lusaka, Zambia
| | - Peter Mwaba
- UNZA-UCLMS Research and Training Project, University Teaching Hospital, Lusaka, Zambia
| | - Klaus Reither
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | - Sarah Edwards
- Ethics and Governance, NIHR BRC at University College London Hospitals NHS Trust, London, United Kingdom
| | | | | | - Pere-Joan Cardona
- Unitat de Tuberculosi Experimental Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i PujolEdifici Laboratoris de Recerca Can Ruti Campus, Barcelona, Spain
| | - James B W Russell
- Department of Internal Medicine, Connaught Hospital, College of Medicine and Allied Health Sciences, University of Sierra Leone, Sierra Leone
| | - Vanya Gant
- Department of Microbiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Mahdad Noursadeghi
- 1] Division of Infection and Immunity at the University College London (UCL), London, UK and National Institute of Health Research's Biomedical Research Centre UCL Hospitals National Health Service Foundation Trust, Royal Free Campus, 2nd Floor, Rowland Hill Street, London NW3 2PF, UK. [2] Department of Microbiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Paul Elkington
- Respiratory Medicine department, Southampton University, Southampton, UK
| | | | - Clara Menendez
- ISGlobal, Barcelona Institute for Global health, Barcelona, Spain. Immunology Unit-Laboratoire Bactériologie Virologie, Le Dantec
| | - Tandakha N Dieye
- Hospital, Laboratories of the National Blood Transfusion Center, Senegal
| | - Bassirou Diarra
- SEREFO (HIV/TB Research and Training CenteHIV/TB Research and Training Center, FMOS, University of STT, Bamako, Mali
| | - Almoustapha Maiga
- SEREFO (HIV/TB Research and Training CenteHIV/TB Research and Training Center, FMOS, University of STT, Bamako, Mali
| | - Abraham Aseffa
- Armaeur Hansen Resrach Institute (AHRI), Addis Ababa, Ethiopia
| | - Shreemanta Parida
- Therapeutic Immunology, Department of Laboratory Medicine, Karolinska Institutet and Center for Allogeneic Stem Cell Transplantation, Karolinska Hospital, Hälsovägen, SE-14186 Stockholm, Sweden
| | - Christian Wejse
- Department of Infectious Diseases, Aarhus University, Aarhus, Denmark
| | - Eskild Petersen
- Department of Infectious Diseases, Aarhus University, Aarhus, Denmark
| | - Pontiano Kaleebu
- Medical Research Council/Uganda Virus Research Institute, Research Unit on AIDS, Entebbe, Uganda
| | | | - Gill Craig
- School of Health Sciences, City University London, City University, London, UK
| | - Tumena Corrah
- Department of Infectious and Tropical Diseases, Northwick Park Hospital, London, UK
| | | | | | - Martin Rao
- Therapeutic Immunology, Department of Laboratory Medicine, Karolinska Institutet and Center for Allogeneic Stem Cell Transplantation, Karolinska Hospital, Hälsovägen, SE-14186 Stockholm, Sweden
| | - Timothy D McHugh
- Division of Infection and Immunity at the University College London (UCL), London, UK and National Institute of Health Research's Biomedical Research Centre UCL Hospitals National Health Service Foundation Trust, Royal Free Campus, 2nd Floor, Rowland Hill Street, London NW3 2PF, UK
| | - Aziz Sheikh
- Centre of Medical Informatics, Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Edinburgh, UK
| | - Giuseppe Ippolito
- National Institute for Infectious Diseases Lazzaro Spallanzani, Rome, Italy
| | - Gita Ramjee
- HIV prevention Research Unit, MRC, Durban South Africa
| | | | | | - Andrie Steyn
- KwaZulu-Natal Research Institute for Tuberculosis &HIV, Nelson R. Mandela School of Medicine, K-RITH Tower Building, Durban, South Africa
| | - Martin Grobusch
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, University of Amsterdam, Amsterdam, The Netherlands
| | - Ian Sanne
- Right to care, University of Witwatersrand, Johannesburg, South Africa
| | | | - Rajhmun Madansein
- Department of Cardiothoracic Surgery, University of KwaZulu Natal, Inkosi Albert Luthuli Central Hospital and King DinuZulu Hospital, Durban, South Africa
| | - Robert J Wilkinson
- Clinical Infectious Diseases Research Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa and Francis Crick Institute Mill Hill Laboratory, London, UK
| | - Bongani Mayosi
- Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa
| | - Marco Schito
- Critical Path to TB Drug Regimens, Critical Path Institute, Tucson, Arizona, USA
| | | |
Collapse
|
186
|
Rubinsztein DC, Bento CF, Deretic V. Therapeutic targeting of autophagy in neurodegenerative and infectious diseases. J Exp Med 2015; 212:979-90. [PMID: 26101267 PMCID: PMC4493419 DOI: 10.1084/jem.20150956] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Autophagy is a conserved process that uses double-membrane vesicles to deliver cytoplasmic contents to lysosomes for degradation. Although autophagy may impact many facets of human biology and disease, in this review we focus on the ability of autophagy to protect against certain neurodegenerative and infectious diseases. Autophagy enhances the clearance of toxic, cytoplasmic, aggregate-prone proteins and infectious agents. The beneficial roles of autophagy can now be extended to supporting cell survival and regulating inflammation. Autophagic control of inflammation is one area where autophagy may have similar benefits for both infectious and neurodegenerative diseases beyond direct removal of the pathogenic agents. Preclinical data supporting the potential therapeutic utility of autophagy modulation in such conditions is accumulating.
Collapse
Affiliation(s)
- David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Cambridge CB2 OSP, England, UK
| | - Carla F Bento
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Cambridge CB2 OSP, England, UK
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology and Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131 Department of Molecular Genetics and Microbiology and Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| |
Collapse
|
187
|
Rana A, Ahmed M, Rub A, Akhter Y. A tug-of-war between the host and the pathogen generates strategic hotspots for the development of novel therapeutic interventions against infectious diseases. Virulence 2015; 6:566-80. [PMID: 26107578 PMCID: PMC4720223 DOI: 10.1080/21505594.2015.1062211] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/02/2015] [Accepted: 06/10/2015] [Indexed: 12/30/2022] Open
Abstract
Microbial pathogens are known to express an array of specific signaling molecules referred as Pathogen Associated Molecular Patterns (PAMPs), which are recognized by Pattern Recognition Receptors (PRRs), present on the surface of the host cells. Interactions between PAMPs and PRRs on the surface of the host cells lead to signaling events which could culminate into either successful infection or clearance of the pathogens. Here, we summarize how these events may generate novel host based as well as pathogen based molecular targets for designing effective therapeutic strategies against infections.
Collapse
Affiliation(s)
- Aarti Rana
- School of Life Sciences; Central University of Himachal Pradesh; Shahpur, District-Kangra, Himachal Pradesh, India
| | - Mushtaq Ahmed
- School of Earth and Environmental Sciences; Central University of Himachal Pradesh; Shahpur, District-Kangra, Himachal Pradesh, India
| | - Abdur Rub
- Infection and Immunity Lab; Department of Biotechnology; Jamia Millia Islamia (A Central University); New Delhi, India
| | - Yusuf Akhter
- School of Life Sciences; Central University of Himachal Pradesh; Shahpur, District-Kangra, Himachal Pradesh, India
| |
Collapse
|
188
|
Abstract
The dramatic rise in microbial drug resistance in recent years has led to ongoing searches for novel drugs to add to the armory against infectious disease. Nevertheless, a paucity of new antibacterial drugs in discovery and development pipelines using traditional approaches has prompted a variety of unconventional and disruptive strategies for antibacterial drug discovery. Herein, we review recent nontraditional approaches that have been piloted for early drug discovery efforts. These unique methodologies open new avenues for finding the next generation of antimicrobials.
Collapse
Affiliation(s)
- Maya A Farha
- M.G. DeGroote Institute for Infectious Disease Research, and Department of Biochemistry and Biomedical Sciences, McMaster University, Ontario, Canada
| | - Eric D Brown
- M.G. DeGroote Institute for Infectious Disease Research, and Department of Biochemistry and Biomedical Sciences, McMaster University, Ontario, Canada
| |
Collapse
|
189
|
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
|
190
|
Espert L, Beaumelle B, Vergne I. Autophagy in Mycobacterium tuberculosis and HIV infections. Front Cell Infect Microbiol 2015; 5:49. [PMID: 26082897 PMCID: PMC4451423 DOI: 10.3389/fcimb.2015.00049] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/18/2015] [Indexed: 12/31/2022] Open
Abstract
Human Immunodeficiency Virus (HIV) and Mycobacterium tuberculosis (M.tb) are among the most lethal human pathogens worldwide, each being responsible for around 1.5 million deaths annually. Moreover, synergy between acquired immune deficiency syndrome (AIDS) and tuberculosis (TB) has turned HIV/M.tb co-infection into a major public health threat in developing countries. In the past decade, autophagy, a lysosomal catabolic process, has emerged as a major host immune defense mechanism against infectious agents like M.tb and HIV. Nevertheless, in some instances, autophagy machinery appears to be instrumental for HIV infection. Finally, there is mounting evidence that both pathogens deploy various countermeasures to thwart autophagy. This mini-review proposes an overview of the roles and regulations of autophagy in HIV and M.tb infections with an emphasis on microbial factors. We also discuss the role of autophagy manipulation in the context of HIV/M.tb co-infection. In future, a comprehensive understanding of autophagy interaction with these pathogens will be critical for development of autophagy-based prophylactic and therapeutic interventions for AIDS and TB.
Collapse
Affiliation(s)
- Lucile Espert
- CPBS FRE 3689 Centre National de la Recherche Scientifique, UM Montpellier, France
| | - Bruno Beaumelle
- CPBS FRE 3689 Centre National de la Recherche Scientifique, UM Montpellier, France
| | - Isabelle Vergne
- Institut de Pharmacologie et de Biologie Structurale, UMR 5089 Centre National de la Recherche Scientifique - Université de Toulouse Toulouse, France
| |
Collapse
|
191
|
Abstract
Host-directed therapies are a relatively new and promising approach to treatment of tuberculosis. Modulation of specific host immune pathways, including those that impact inflammation and immunopathology, can limit mycobacterial infection and pathology, both in cell culture and in animal models. This review explores a range of host pathways and drugs, some already approved for clinical use that have the potential to provide new adjunctive therapies for tuberculosis. Drugs targeting host processes may largely avoid the development of bacterial antibiotic resistance, a major public health concern for tuberculosis. However, these drugs may also have generally increased risk for side effects on the host. Understanding the specific mechanisms by which these drugs act and the relationship of these mechanisms to Mycobacterium tuberculosis pathogenesis will be critical in selecting appropriate host-directed therapy. Overall, these host-directed compounds provide a novel strategy for antituberculosis therapy.
Collapse
Affiliation(s)
- David M Tobin
- Department of Molecular Genetics and Microbiology, Center for Microbial Pathogenesis, Center for AIDS Research, Duke University School of Medicine, Durham, North Carolina 27710
| |
Collapse
|
192
|
Abstract
Tuberculosis caused by Mycobacterium tuberculosis is a global health emergency. This deadly disease has far-reaching social and economic implications. Diseased individuals need prolonged polypharmacy which is not without ill effects. Treatment compliance is often compromised contributing to rising resistance. HIV co-infection has further worsened the scenario. On the other hand, no new anti-TB drug has hit the market in last 4–5 decades. After a long latency, only the last few years have witnessed growing research in this direction and a widening anti-TB drug clinical pipeline. The compounds in preclinical stage of development have also shown a heartening increase. The present review is an attempt to discuss novel promising patents in this field.
Collapse
|
193
|
Arreola R, Becerril-Villanueva E, Cruz-Fuentes C, Velasco-Velázquez MA, Garcés-Alvarez ME, Hurtado-Alvarado G, Quintero-Fabian S, Pavón L. Immunomodulatory effects mediated by serotonin. J Immunol Res 2015; 2015:354957. [PMID: 25961058 PMCID: PMC4417587 DOI: 10.1155/2015/354957] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 02/24/2015] [Indexed: 11/17/2022] Open
Abstract
Serotonin (5-HT) induces concentration-dependent metabolic effects in diverse cell types, including neurons, entherochromaffin cells, adipocytes, pancreatic beta-cells, fibroblasts, smooth muscle cells, epithelial cells, and leukocytes. Three classes of genes regulating 5-HT function are constitutively expressed or induced in these cells: (a) membrane proteins that regulate the response to 5-HT, such as SERT, 5HTR-GPCR, and the 5HT3-ion channels; (b) downstream signaling transduction proteins; and (c) enzymes controlling 5-HT metabolism, such as IDO and MAO, which can generate biologically active catabolites, including melatonin, kynurenines, and kynurenamines. This review covers the clinical and experimental mechanisms involved in 5-HT-induced immunomodulation. These mechanisms are cell-specific and depend on the expression of serotonergic components in immune cells. Consequently, 5-HT can modulate several immunological events, such as chemotaxis, leukocyte activation, proliferation, cytokine secretion, anergy, and apoptosis. The effects of 5-HT on immune cells may be relevant in the clinical outcome of pathologies with an inflammatory component. Major depression, fibromyalgia, Alzheimer disease, psoriasis, arthritis, allergies, and asthma are all associated with changes in the serotonergic system associated with leukocytes. Thus, pharmacological regulation of the serotonergic system may modulate immune function and provide therapeutic alternatives for these diseases.
Collapse
Affiliation(s)
- Rodrigo Arreola
- Psychiatric Genetics Department, Clinical Research Branch, National Institute of Psychiatry, “Ramón de la Fuente”, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, DF, Mexico
| | - Enrique Becerril-Villanueva
- Department of Psychoimmunology, National Institute of Psychiatry, “Ramón de la Fuente”, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, DF, Mexico
| | - Carlos Cruz-Fuentes
- Psychiatric Genetics Department, Clinical Research Branch, National Institute of Psychiatry, “Ramón de la Fuente”, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, DF, Mexico
| | - Marco Antonio Velasco-Velázquez
- School of Medicine, National Autonomous University of Mexico, Avenida Universidad 3000, Coyoacan, 04510 Mexico City, DF, Mexico
| | - María Eugenia Garcés-Alvarez
- Department of Psychoimmunology, National Institute of Psychiatry, “Ramón de la Fuente”, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, DF, Mexico
| | - Gabriela Hurtado-Alvarado
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autonoma Metropolitana, Unidad Iztapalapa, Avenida San Rafael Atlixco No. 186, Colonia Vicentina, Iztapalapa, 09340 Mexico City, DF, Mexico
| | - Saray Quintero-Fabian
- Genetics Unit Nutrition of Biomedical Research Institute of Universidad Nacional Autónoma de México at Instituto Nacional de Pediatría, Avenida del Iman No. 1, cuarto piso, Colonia Insurgentes-Cuicuilco, Coyoacan, 04530 Mexico City, DF, Mexico
| | - Lenin Pavón
- Department of Psychoimmunology, National Institute of Psychiatry, “Ramón de la Fuente”, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, DF, Mexico
| |
Collapse
|
194
|
Julián E, Baelo A, Gavaldà J, Torrents E. Methyl-hydroxylamine as an efficacious antibacterial agent that targets the ribonucleotide reductase enzyme. PLoS One 2015; 10:e0122049. [PMID: 25782003 PMCID: PMC4363900 DOI: 10.1371/journal.pone.0122049] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 02/06/2015] [Indexed: 01/17/2023] Open
Abstract
The emergence of multidrug-resistant bacteria has encouraged vigorous efforts to develop antimicrobial agents with new mechanisms of action. Ribonucleotide reductase (RNR) is a key enzyme in DNA replication that acts by converting ribonucleotides into the corresponding deoxyribonucleotides, which are the building blocks of DNA replication and repair. RNR has been extensively studied as an ideal target for DNA inhibition, and several drugs that are already available on the market are used for anticancer and antiviral activity. However, the high toxicity of these current drugs to eukaryotic cells does not permit their use as antibacterial agents. Here, we present a radical scavenger compound that inhibited bacterial RNR, and the compound's activity as an antibacterial agent together with its toxicity in eukaryotic cells were evaluated. First, the efficacy of N-methyl-hydroxylamine (M-HA) in inhibiting the growth of different Gram-positive and Gram-negative bacteria was demonstrated, and no effect on eukaryotic cells was observed. M-HA showed remarkable efficacy against Mycobacterium bovis BCG and Pseudomonas aeruginosa. Thus, given the M-HA activity against these two bacteria, our results showed that M-HA has intracellular antimycobacterial activity against BCG-infected macrophages, and it is efficacious in partially disassembling and inhibiting the further formation of P. aeruginosa biofilms. Furthermore, M-HA and ciprofloxacin showed a synergistic effect that caused a massive reduction in a P. aeruginosa biofilm. Overall, our results suggest the vast potential of M-HA as an antibacterial agent, which acts by specifically targeting a bacterial RNR enzyme.
Collapse
Affiliation(s)
- Esther Julián
- Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Aida Baelo
- Institute for Bioengineering of Catalonia (IBEC), Bacterial infections and antimicrobial therapies; Baldiri Reixac 15-21, Barcelona, Spain
| | - Joan Gavaldà
- Infectious Diseases Research Laboratory, Infectious Diseases Department, Vall d’Hebron Research Institute, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Eduard Torrents
- Institute for Bioengineering of Catalonia (IBEC), Bacterial infections and antimicrobial therapies; Baldiri Reixac 15-21, Barcelona, Spain
- * E-mail:
| |
Collapse
|
195
|
Abstract
Improved treatments are needed for nearly all forms of Mycobacterium tuberculosis infection. Adjunctive host-directed therapies have the potential to shorten tuberculosis treatment duration, prevent resistance and reduce lung injury by promoting autophagy, antimicrobial peptide production and other macrophage effector mechanisms, as well as by modifying specific mechanisms that cause lung inflammation and matrix destruction. The range of candidates is broad, including several agents approved for other clinical indications that are ready for evaluation in Phase II clinical trials. The promise of new and existing host-directed therapies that could accelerate response and improve tuberculosis treatment outcomes is discussed in this Opinion article.
Collapse
|
196
|
Durairaj H, Steury MD, Parameswaran N. Paroxetine differentially modulates LPS-induced TNFα and IL-6 production in mouse macrophages. Int Immunopharmacol 2015; 25:485-92. [PMID: 25744603 DOI: 10.1016/j.intimp.2015.02.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 02/10/2015] [Accepted: 02/18/2015] [Indexed: 11/16/2022]
Abstract
Paroxetine is a selective serotonin reuptake inhibitor (SSRI) that is clinically used for the treatment of depression in human patients. Because of recent reports on the role of serotonin in modulating inflammation and the link between inflammation and depression, we sought to test the effect of paroxetine directly on macrophage response to an inflammatory stimulus. Lipopolysaccharide (LPS) treatment of mouse macrophages significantly enhanced TNFα and IL-6 production. Paroxetine treatment of macrophages, however, significantly inhibited LPS-induced IL-6 production. In contrast, paroxetine enhanced LPS-induced TNFα production in macrophages. These effects of paroxetine were mimicked by fluoxetine, another SSRI. To determine if the effects of paroxetine are mediated via modulation of the 5-HT system, we treated macrophages with 5-HT or 5-HT receptor antagonist (LY215840) in the presence of LPS and/or paroxetine. 5-HT treatment by itself did not affect LPS-induced cytokine production. LY215840, however, reversed paroxetine's effect on LPS-induced TNFα production but not IL-6. To understand the signaling mechanisms, we examined paroxetine's effect on MAPK and NFκB pathways. While paroxetine inhibited LPS-induced IκBα phosphorylation, MAPK pathways were mostly unaffected. Together these data demonstrate that paroxetine has critical but differential effects on IL-6 and TNFα production in macrophages and that it likely regulates these cytokines via distinct mechanisms.
Collapse
Affiliation(s)
- Haritha Durairaj
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Michael D Steury
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | | |
Collapse
|
197
|
Abstract
Macrophages and neutrophils play a decisive role in host responses to intracellular bacteria including the agent of tuberculosis (TB), Mycobacterium tuberculosis as they represent the forefront of innate immune defense against bacterial invaders. At the same time, these phagocytes are also primary targets of intracellular bacteria to be abused as host cells. Their efficacy to contain and eliminate intracellular M. tuberculosis decides whether a patient initially becomes infected or not. However, when the infection becomes chronic or even latent (as in the case of TB) despite development of specific immune activation, phagocytes have also important effector functions. Macrophages have evolved a myriad of defense strategies to combat infection with intracellular bacteria such as M. tuberculosis. These include induction of toxic anti-microbial effectors such as nitric oxide and reactive oxygen intermediates, the stimulation of microbe intoxication mechanisms via acidification or metal accumulation in the phagolysosome, the restriction of the microbe's access to essential nutrients such as iron, fatty acids, or amino acids, the production of anti-microbial peptides and cytokines, along with induction of autophagy and efferocytosis to eliminate the pathogen. On the other hand, M. tuberculosis, as a prime example of a well-adapted facultative intracellular bacterium, has learned during evolution to counter-balance the host's immune defense strategies to secure survival or multiplication within this otherwise hostile environment. This review provides an overview of innate immune defense of macrophages directed against intracellular bacteria with a focus on M. tuberculosis. Gaining more insights and knowledge into this complex network of host-pathogen interaction will identify novel target sites of intervention to successfully clear infection at a time of rapidly emerging multi-resistance of M. tuberculosis against conventional antibiotics.
Collapse
Affiliation(s)
- Günter Weiss
- Department of Internal Medicine VI, Infectious Disease, Immunology, Rheumatology, Pneumology, Medical University of InnsbruckInnsbruck, Austria
| | - Ulrich E Schaible
- Cellular Microbiology, Priority Area Infections, Research Center BorstelBorstel, Germany
- Department of Immunology, London School of Hygiene and Tropical MedicineLondon, UK
- German Centre of Infection Research, TTU-TBBorstel, Germany
| |
Collapse
|
198
|
Bento CF, Empadinhas N, Mendes V. Autophagy in the fight against tuberculosis. DNA Cell Biol 2015; 34:228-42. [PMID: 25607549 DOI: 10.1089/dna.2014.2745] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tuberculosis (TB), a chronic infectious disease mainly caused by the tubercle bacillus Mycobacterium tuberculosis, is one of the world's deadliest diseases that has afflicted humanity since ancient times. Although the number of people falling ill with TB each year is declining, its incidence in many developing countries is still a major cause of concern. Upon invading host cells by phagocytosis, M. tuberculosis can replicate within infected cells by arresting the maturation of the phagosome whose function is to target the pathogen for elimination. Host cells have mechanisms of controlling this evasion by inducing autophagy, an elaborate cellular process that targets bacteria for progressive elimination, decreasing bacterial loads within infected cells. In addition, autophagy activation also aids in the control of inflammation, contributing to a more efficient innate immune response against M. tuberculosis. Several innovative TB therapies have been envisaged based on autophagy manipulation, with some of them revealing high potential for future clinical trials and eventual implementation in healthcare systems. Thus, this review highlights the recent advances on the innate immune response regulation by autophagy upon M. tuberculosis infection and the promising new autophagy-based therapies for TB.
Collapse
Affiliation(s)
- Carla F Bento
- 1 Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge , Cambridge, United Kingdom
| | | | | |
Collapse
|
199
|
Fu YR, Gao KS, Ji R, Yi ZJ. Differential transcriptional response in macrophages infected with cell wall deficient versus normal Mycobacterium Tuberculosis. Int J Biol Sci 2015; 11:22-30. [PMID: 25552926 PMCID: PMC4278251 DOI: 10.7150/ijbs.10217] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/13/2014] [Indexed: 02/06/2023] Open
Abstract
Host-pathogen interactions determine the outcome following infection by mycobacterium tuberculosis (Mtb). Under adverse circumstances, normal Mtb can form cell-wall deficient (CWD) variants within macrophages, which have been considered an adaptive strategy for facilitating bacterial survival inside macrophages. However, the molecular mechanism by which infection of macrophages with different phenotypic Mtb elicits distinct responses of macrophages is not fully understood. To explore the molecular events triggered upon Mtb infection of macrophages, differential transcriptional responses of RAW264.7 cells infected with two forms of Mtb, CWD-Mtb and normal Mtb, were studied by microarray analysis. Some of the differentially regulated genes were confirmed by RT-qPCR in both RAW264.7 cells and primary macrophages. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway was used to analyze functions of differentially expressed genes. Distinct gene expression patterns were observed between CWD-Mtb and normal Mtb group. Mapt was up-regulated, while NOS2 and IL-11 were down-regulated in CWD-Mtb infected RAW264.7 cells and primary macrophages compared with normal Mtb infected ones. Many deregulated genes were found to be related to macrophages activation, immune response, phagosome maturation, autophagy and lipid metabolism. KEGG analysis showed that the differentially expressed genes were mainly involved in MAPK signaling pathway, nitrogen metabolism, cytokine-cytokine receptor interaction and focal adhesion. Taken together, the present study showed that differential macrophage responses were induced by intracellular CWD-Mtb an normal Mtb infection, which suggested that interactions between macrophages and different phenotypic Mtb are very complex. The results provide evidence for further understanding of pathogenesis of CWD-Mtb and may help in improving strategies to eliminate intracellular CWD-Mtb.
Collapse
Affiliation(s)
- Yu-Rong Fu
- 1. Department of Laboratory Medicine of Affiliated Hospital of Weifang Medical University, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong and Medical Priority Speciality of Clinical Laboratory in Shandong Province, Weifang Medical University, Weifang 261031, China; ; 2. Department of Medical Microbiology, Weifang Medical University, Weifang 261053, China
| | - Kun-Shan Gao
- 1. Department of Laboratory Medicine of Affiliated Hospital of Weifang Medical University, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong and Medical Priority Speciality of Clinical Laboratory in Shandong Province, Weifang Medical University, Weifang 261031, China
| | - Rui Ji
- 2. Department of Medical Microbiology, Weifang Medical University, Weifang 261053, China
| | - Zheng-Jun Yi
- 1. Department of Laboratory Medicine of Affiliated Hospital of Weifang Medical University, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong and Medical Priority Speciality of Clinical Laboratory in Shandong Province, Weifang Medical University, Weifang 261031, China
| |
Collapse
|
200
|
Delorme V, Song OR, Baulard A, Brodin P. Testing chemical and genetic Modulators in Mycobacterium tuberculosis infected cells using phenotypic assays. Methods Mol Biol 2015; 1285:387-411. [PMID: 25779330 DOI: 10.1007/978-1-4939-2450-9_24] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Mycobacterium tuberculosis is able to colonize host cells, and it is now well admitted that the intracellular stage of the bacteria contributes to tuberculosis pathogenesis as well as to making it a persistent infection. There is still limited understanding on how the tubercle bacillus colonizes the cell and what are the factors impacting on its intracellular persistence. Recent advances in imaging technique allow rapid quantification of biological objects in complex environments. Furthermore, M. tuberculosis is a microorganism that is particularly genetically tractable and that tolerates the expression of heterologous fluorescent proteins. Thus, the intracellular distribution of M. tuberculosis expressing fluorescent proteins can be easily quantified by the use of confocal microscopy. Here we describe high-content/high-throughput imaging methods that enable tracking the bacillus inside host settings, taking into account the heterogeneity of colonization.
Collapse
Affiliation(s)
- Vincent Delorme
- Institut Pasteur de Lille, University of Lille Inserm U1019, CNRS UMR8204, Institut Pasteur de Lille, 1, rue du Professeur Calmette, BP245, Lille Cedex, 59019, France
| | | | | | | |
Collapse
|