1
|
Aiello A, Najafi-Fard S, Goletti D. Initial immune response after exposure to Mycobacterium tuberculosis or to SARS-COV-2: similarities and differences. Front Immunol 2023; 14:1244556. [PMID: 37662901 PMCID: PMC10470049 DOI: 10.3389/fimmu.2023.1244556] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) and Coronavirus disease-2019 (COVID-19), whose etiologic agent is severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), are currently the two deadliest infectious diseases in humans, which together have caused about more than 11 million deaths worldwide in the past 3 years. TB and COVID-19 share several aspects including the droplet- and aerosol-borne transmissibility, the lungs as primary target, some symptoms, and diagnostic tools. However, these two infectious diseases differ in other aspects as their incubation period, immune cells involved, persistence and the immunopathological response. In this review, we highlight the similarities and differences between TB and COVID-19 focusing on the innate and adaptive immune response induced after the exposure to Mtb and SARS-CoV-2 and the pathological pathways linking the two infections. Moreover, we provide a brief overview of the immune response in case of TB-COVID-19 co-infection highlighting the similarities and differences of each individual infection. A comprehensive understanding of the immune response involved in TB and COVID-19 is of utmost importance for the design of effective therapeutic strategies and vaccines for both diseases.
Collapse
Affiliation(s)
| | | | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
2
|
Mycobacterium tuberculosis Dormancy: How to Fight a Hidden Danger. Microorganisms 2022; 10:microorganisms10122334. [PMID: 36557586 PMCID: PMC9784227 DOI: 10.3390/microorganisms10122334] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Both latent and active TB infections are caused by a heterogeneous population of mycobacteria, which includes actively replicating and dormant bacilli in different proportions. Dormancy substantially affects M. tuberculosis drug tolerance and TB clinical management due to a significant decrease in the metabolic activity of bacilli, which leads to the complexity of both the diagnosis and the eradication of bacilli. Most diagnostic approaches to latent infection deal with a subpopulation of active M. tuberculosis, underestimating the contribution of dormant bacilli and leading to limited success in the fight against latent TB. Moreover, active TB appears not only as a primary form of infection but can also develop from latent TB, when resuscitation from dormancy is followed by bacterial multiplication, leading to disease progression. To win against latent infection, the identification of the Achilles' heel of dormant M. tuberculosis is urgently needed. Regulatory mechanisms and metabolic adaptation to growth arrest should be studied using in vitro and in vivo models that adequately imitate latent TB infection in macroorganisms. Understanding the mechanisms underlying M. tuberculosis dormancy and resuscitation may provide clues to help control latent infection, reduce disease severity in patients, and prevent pathogen transmission in the population.
Collapse
|
3
|
Ncube P, Bagheri B, Goosen WJ, Miller MA, Sampson SL. Evidence, Challenges, and Knowledge Gaps Regarding Latent Tuberculosis in Animals. Microorganisms 2022; 10:1845. [PMID: 36144447 PMCID: PMC9503773 DOI: 10.3390/microorganisms10091845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 01/30/2023] Open
Abstract
Mycobacterium bovis and other Mycobacterium tuberculosis complex (MTBC) pathogens that cause domestic animal and wildlife tuberculosis have received considerably less attention than M. tuberculosis, the primary cause of human tuberculosis (TB). Human TB studies have shown that different stages of infection can exist, driven by host-pathogen interactions. This results in the emergence of heterogeneous subpopulations of mycobacteria in different phenotypic states, which range from actively replicating (AR) cells to viable but slowly or non-replicating (VBNR), viable but non-culturable (VBNC), and dormant mycobacteria. The VBNR, VBNC, and dormant subpopulations are believed to underlie latent tuberculosis (LTB) in humans; however, it is unclear if a similar phenomenon could be happening in animals. This review discusses the evidence, challenges, and knowledge gaps regarding LTB in animals, and possible host-pathogen differences in the MTBC strains M. tuberculosis and M. bovis during infection. We further consider models that might be adapted from human TB research to investigate how the different phenotypic states of bacteria could influence TB stages in animals. In addition, we explore potential host biomarkers and mycobacterial changes in the DosR regulon, transcriptional sigma factors, and resuscitation-promoting factors that may influence the development of LTB.
Collapse
Affiliation(s)
| | | | | | | | - Samantha Leigh Sampson
- DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Department of Biomedical Sciences, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Francie Van Zijl Dr, Parow, Cape Town 7505, South Africa
| |
Collapse
|
4
|
Bouzeyen R, Chugh S, Gosain TP, Barbouche MR, Haoues M, Rao KVS, Essafi M, Singh R. Co-Administration of Anticancer Candidate MK-2206 Enhances the Efficacy of BCG Vaccine Against Mycobacterium tuberculosis in Mice and Guinea Pigs. Front Immunol 2021; 12:645962. [PMID: 34122406 PMCID: PMC8190480 DOI: 10.3389/fimmu.2021.645962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/29/2021] [Indexed: 01/19/2023] Open
Abstract
The failure of M. bovis BCG to induce long-term protection has been endowed to its inability to escape the phagolysosome, leading to mild activation of CD8+ mediated T cell response. Induction of apoptosis in host cells plays an important role in potentiating dendritic cells-mediated priming of CD8+ T cells, a process defined as “cross-priming.” Moreover, IL-10 secretion by infected cells has been reported to hamper BCG-induced immunity against Tuberculosis (TB). Previously, we have reported that apoptosis of BCG-infected macrophages and inhibition of IL-10 secretion is FOXO3 dependent, a transcription factor negatively regulated by the pro-survival activated threonine kinase, Akt. We speculate that FOXO3-mediated induction of apoptosis and abrogation of IL-10 secretion along with M. bovis BCG immunization might enhance the protection imparted by BCG. Here, we have assessed whether co-administration of a known anti-cancer Akt inhibitor, MK-2206, enhances the protective efficacy of M. bovis BCG in mice model of infection. We observed that in vitro MK-2206 treatment resulted in FOXO3 activation, enhanced BCG-induced apoptosis of macrophages and inhibition of IL-10 secretion. Co-administration of M. bovis BCG along with MK-2206 also increased apoptosis of antigen-presenting cells in draining lymph nodes of immunized mice. Further, MK-2206 administration improved BCG-induced CD4+ and CD8+ effector T cells responses and its ability to induce both effector and central memory T cells. Finally, we show that co-administration of MK-2206 enhanced the protection imparted by M. bovis BCG against Mtb in aerosol infected mice and guinea pigs. Taken together, we provide evidence that MK-2206-mediated activation of FOXO3 potentiates BCG-induced immunity and imparts protection against Mtb through enhanced innate immune response.
Collapse
Affiliation(s)
- Rania Bouzeyen
- Institut Pasteur de Tunis, LTCII, LR11 IPT02, Tunis, Tunisia
| | - Saurabh Chugh
- Translational Health Science and Technology Institute, Faridabad, India
| | | | | | - Meriam Haoues
- Institut Pasteur de Tunis, LTCII, LR11 IPT02, Tunis, Tunisia
| | - Kanury V S Rao
- Translational Health Science and Technology Institute, Faridabad, India
| | - Makram Essafi
- Institut Pasteur de Tunis, LTCII, LR11 IPT02, Tunis, Tunisia
| | - Ramandeep Singh
- Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
5
|
Ardiaca García M, Montesinos Barceló A, Bonvehí Nadeu C, Jekl V. Respiratory Diseases in Guinea Pigs, Chinchillas and Degus. Vet Clin North Am Exot Anim Pract 2021; 24:419-457. [PMID: 33892894 DOI: 10.1016/j.cvex.2021.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The diagnosis and treatment of respiratory disease in pet guinea pigs, chinchillas, and degus still face profoundly serious challenges owing to their relatively small size, conspicuous clinical signs, difficulty for sampling, and insufficient scientific evidence to correlate signs and particular pathologies. This article is intended to summarize the available information on the relevant anatomy, physiology, and respiratory pathology in these species.
Collapse
Affiliation(s)
| | - Andrés Montesinos Barceló
- Centro Veterinario Los Sauces, Calle Santa Engracia, 63, Madrid 28010, Spain; Department of Animal Medicine and Surgery, Veterinary Faculty, Universidad Complutense de Madrid, Avenida Puerta de Hierro, s/n 28040 Madrid Spain
| | | | - Vladimír Jekl
- Small Mammal, Department of Pharmacology and Pharmacy, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic; Jekl and Hauptman Veterinary Clinic, Focused on Exotic Companion Mammal Care, Mojmírovo Náměstí 3105/6a, Brno 612 00, Czech Republic
| |
Collapse
|
6
|
Campaniço A, Harjivan SG, Warner DF, Moreira R, Lopes F. Addressing Latent Tuberculosis: New Advances in Mimicking the Disease, Discovering Key Targets, and Designing Hit Compounds. Int J Mol Sci 2020; 21:ijms21228854. [PMID: 33238468 PMCID: PMC7700174 DOI: 10.3390/ijms21228854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
Despite being discovered and isolated more than one hundred years ago, tuberculosis (TB) remains a global public health concern arch. Our inability to eradicate this bacillus is strongly related with the growing resistance, low compliance to current drugs, and the capacity of the bacteria to coexist in a state of asymptomatic latency. This last state can be sustained for years or even decades, waiting for a breach in the immune system to become active again. Furthermore, most current therapies are not efficacious against this state, failing to completely clear the infection. Over the years, a series of experimental methods have been developed to mimic the latent state, currently used in drug discovery, both in vitro and in vivo. Most of these methods focus in one specific latency inducing factor, with only a few taking into consideration the complexity of the granuloma and the genomic and proteomic consequences of each physiological factor. A series of targets specifically involved in latency have been studied over the years with promising scaffolds being discovered and explored. Taking in account that solving the latency problem is one of the keys to eradicate the disease, herein we compile current therapies and diagnosis techniques, methods to mimic latency and new targets and compounds in the pipeline of drug discovery.
Collapse
Affiliation(s)
- André Campaniço
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Shrika G. Harjivan
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Digby F. Warner
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa;
- Department of Pathology, SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, University of Cape Town, Rondebosch 7701, South Africa
- Welcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch 7701, South Africa
| | - Rui Moreira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Francisca Lopes
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
- Correspondence:
| |
Collapse
|
7
|
Li J, Zhao A, Tang J, Wang G, Shi Y, Zhan L, Qin C. Tuberculosis vaccine development: from classic to clinical candidates. Eur J Clin Microbiol Infect Dis 2020; 39:1405-1425. [PMID: 32060754 PMCID: PMC7223099 DOI: 10.1007/s10096-020-03843-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/05/2020] [Indexed: 12/12/2022]
Abstract
Bacillus Calmette-Guérin (BCG) has been in use for nearly 100 years and is the only licensed TB vaccine. While BCG provides protection against disseminated TB in infants, its protection against adult pulmonary tuberculosis (PTB) is variable. To achieve the ambitious goal of eradicating TB worldwide by 2050, there is an urgent need to develop novel TB vaccines. Currently, there are more than a dozen novel TB vaccines including prophylactic and therapeutic at different stages of clinical research. This literature review provides an overview of the clinical status of candidate TB vaccines and discusses the challenges and future development trends of novel TB vaccine research in combination with the efficacy of evaluation of TB vaccines, provides insight for the development of safer and more efficient vaccines, and may inspire new ideas for the prevention of TB.
Collapse
Affiliation(s)
- Junli Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Aihua Zhao
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, People's Republic of China
| | - Jun Tang
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Guozhi Wang
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, People's Republic of China
| | - Yanan Shi
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Lingjun Zhan
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China.
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China.
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China.
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China.
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China.
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China.
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China.
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China.
| |
Collapse
|
8
|
Gao J, Guo M, Teng L, Bao R, Xian Q, Wang X, Ho W. Guinea pig infected with Mycobacterium tuberculosis via oral consumption. JOURNAL OF APPLIED ANIMAL RESEARCH 2018. [DOI: 10.1080/09712119.2018.1505622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Jianfeng Gao
- School of Basic Medical Sciences, Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan, People’s Republic of China
| | - Ming Guo
- School of Basic Medical Sciences, Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan, People’s Republic of China
| | - Li Teng
- Department of Pathology, Wuhan Medical Care Center for Women and Children, Wuhan, People’s Republic of China
| | - Rong Bao
- School of Basic Medical Sciences, Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan, People’s Republic of China
| | - Qiaoyang Xian
- School of Basic Medical Sciences, Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan, People’s Republic of China
| | - Xin Wang
- School of Basic Medical Sciences, Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan, People’s Republic of China
| | - Wenzhe Ho
- School of Basic Medical Sciences, Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan, People’s Republic of China
| |
Collapse
|
9
|
Sousa EHS, Gonzalez G, Gilles-Gonzalez MA. Target DNA stabilizes Mycobacterium tuberculosis DevR/DosR phosphorylation by the full-length oxygen sensors DevS/DosS and DosT. FEBS J 2017; 284:3954-3967. [PMID: 28977726 DOI: 10.1111/febs.14284] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/14/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022]
Abstract
Mycobacterium tuberculosis strongly relies on a latency, or nonreplicating persistence, to escape a human host's immune system. The DevR (DosR), DevS (DosS), and DosT proteins are key components of this process. Like the rhizobial FixL oxygen sensor, DevS and DosT are histidine protein kinases with a heme-binding domain. Like the FixJ partner and substrate of FixL, DevR is a classical response regulator of the two-component class. When activated by DevS or DosT during hypoxia in vivo, DevR induces a dormancy regulon of more than 40 genes. To investigate the contributions of DevS, DosT, and target DNA to the phosphorylation of DevR, we developed an in vitro assay in which the full-length, sensing, DevS and DosT proteins were used to phosphorylate DevR with ATP, in the presence of target DNAs that were introduced as oligonucleotides linked to magnetic nanoparticles. We found that the DevR phosphorylations proceeded only for the deoxy states of the sensors. The reaction was strongly inhibited by O2 , but not CO or NO. The production of phospho-DevR was enhanced sixfold by target consensus DNA or acr-DNA. The phospho-DevR bound tightly to that DNA (Kd ~ 0.8 nm toward acr-DNA), and it was only slightly displaced by a 200-fold excess of unphosphorylated DevR or of a truncated DevR with only a DNA-binding domain. To our knowledge, this represents the first in vitro study of the ligand regulation of DevR phosphorylation by full-length DevS and DosT, and demonstration of a positive effect of DNA on this reaction.
Collapse
Affiliation(s)
- Eduardo H S Sousa
- Laboratory of Bioinorganic Chemistry, Department of Organic and Inorganic Chemistry, Federal University of Ceara, Center for Sciences, Fortaleza, Brazil
| | - Gonzalo Gonzalez
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
10
|
Zhan L, Tang J, Sun M, Qin C. Animal Models for Tuberculosis in Translational and Precision Medicine. Front Microbiol 2017; 8:717. [PMID: 28522990 PMCID: PMC5415616 DOI: 10.3389/fmicb.2017.00717] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/06/2017] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB) is a health threat to the global population. Anti-TB drugs and vaccines are key approaches for TB prevention and control. TB animal models are basic tools for developing biomarkers of diagnosis, drugs for therapy, vaccines for prevention and researching pathogenic mechanisms for identification of targets; thus, they serve as the cornerstone of comparative medicine, translational medicine, and precision medicine. In this review, we discuss the current use of TB animal models and their problems, as well as offering perspectives on the future of these models.
Collapse
Affiliation(s)
- Lingjun Zhan
- Key Laboratory of Human Disease Comparative Medicine, Ministry of HealthBeijing, China.,Institution of Laboratory Animal Sciences, Centre for Tuberculosis, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China.,Beijing Key Laboratory for Animal Models of Emerging and Reemerging InfectiousBeijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijing, China.,Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese MedicineBeijing, China
| | - Jun Tang
- Key Laboratory of Human Disease Comparative Medicine, Ministry of HealthBeijing, China.,Institution of Laboratory Animal Sciences, Centre for Tuberculosis, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China.,Beijing Key Laboratory for Animal Models of Emerging and Reemerging InfectiousBeijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijing, China.,Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese MedicineBeijing, China
| | - Mengmeng Sun
- Key Laboratory of Human Disease Comparative Medicine, Ministry of HealthBeijing, China.,Institution of Laboratory Animal Sciences, Centre for Tuberculosis, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China.,Beijing Key Laboratory for Animal Models of Emerging and Reemerging InfectiousBeijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijing, China.,Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese MedicineBeijing, China
| | - Chuan Qin
- Key Laboratory of Human Disease Comparative Medicine, Ministry of HealthBeijing, China.,Institution of Laboratory Animal Sciences, Centre for Tuberculosis, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China.,Beijing Key Laboratory for Animal Models of Emerging and Reemerging InfectiousBeijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijing, China.,Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese MedicineBeijing, China
| |
Collapse
|
11
|
Sebastian J, Swaminath S, Nair RR, Jakkala K, Pradhan A, Ajitkumar P. De Novo Emergence of Genetically Resistant Mutants of Mycobacterium tuberculosis from the Persistence Phase Cells Formed against Antituberculosis Drugs In Vitro. Antimicrob Agents Chemother 2017; 61:e01343-16. [PMID: 27895008 PMCID: PMC5278719 DOI: 10.1128/aac.01343-16] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 11/16/2016] [Indexed: 12/19/2022] Open
Abstract
Bacterial persisters are a subpopulation of cells that can tolerate lethal concentrations of antibiotics. However, the possibility of the emergence of genetically resistant mutants from antibiotic persister cell populations, upon continued exposure to lethal concentrations of antibiotics, remained unexplored. In the present study, we found that Mycobacterium tuberculosis cells exposed continuously to lethal concentrations of rifampin (RIF) or moxifloxacin (MXF) for prolonged durations showed killing, RIF/MXF persistence, and regrowth phases. RIF-resistant or MXF-resistant mutants carrying clinically relevant mutations in the rpoB or gyrA gene, respectively, were found to emerge at high frequency from the RIF persistence phase population. A Luria-Delbruck fluctuation experiment using RIF-exposed M. tuberculosis cells showed that the rpoB mutants were not preexistent in the population but were formed de novo from the RIF persistence phase population. The RIF persistence phase M. tuberculosis cells carried elevated levels of hydroxyl radical that inflicted extensive genome-wide mutations, generating RIF-resistant mutants. Consistent with the elevated levels of hydroxyl radical-mediated genome-wide random mutagenesis, MXF-resistant M. tuberculosis gyrA de novo mutants could be selected from the RIF persistence phase cells. Thus, unlike previous studies, which showed emergence of genetically resistant mutants upon exposure of bacteria for short durations to sublethal concentrations of antibiotics, our study demonstrates that continuous prolonged exposure of M. tuberculosis cells to lethal concentrations of an antibiotic generates antibiotic persistence phase cells that form a reservoir for the generation of genetically resistant mutants to the same antibiotic or another antibiotic. These findings may have clinical significance in the emergence of drug-resistant tubercle bacilli.
Collapse
Affiliation(s)
- Jees Sebastian
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Sharmada Swaminath
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Rashmi Ravindran Nair
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Kishor Jakkala
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Atul Pradhan
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Parthasarathi Ajitkumar
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| |
Collapse
|
12
|
Ma L, Yang J, Yang L, Shi J, Xue J, Li Y, Liu X. Developmental expression of Toll‑like receptors in the guinea pig lung. Mol Med Rep 2017; 15:1243-1251. [PMID: 28098883 PMCID: PMC5367368 DOI: 10.3892/mmr.2017.6129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 11/21/2016] [Indexed: 12/27/2022] Open
Abstract
The guinea pig is a useful model for investigating infectious and non‑infectious lung diseases due to the sensitivity of its respiratory system and susceptibility to infectious agents. Toll‑like receptors (TLRs) are important components of the innate immune response and are critical for lung immune function. In the present study, the differentiation of epithelial cells in the guinea pig lung was examined during gestation by studying anatomic morphology and the major epithelial cell types using cell type‑specific markers. The developmental expression of all 9 TLRs and the TLR signaling adaptors myeloid differentiation factor 88 (MyD88) and tumor necrosis factor receptor associated factor 6 (TRAF‑6) were investigated by reverse transcription‑quantitative polymerase chain reaction and western blotting analysis. The formation of lung lobes in guinea pigs was observed at 45 days of gestation (dGA), along with the expression of the basal cell marker keratin 14 and the alveolar type II cell marker pro‑surfactant protein. However, the cube cell marker secretoglobin family1A member 1 and ciliated cell marker b‑tubulin IV were only detected in the lungs from 52 dGA onward. The expression levels of all TLRs, MyD88 and TRAF‑6 were determined in lung tissues harvested from embryos, newborn, postnatal and adult animals. The expression levels of all TLR signaling components displayed similar dynamic expression patterns with gestation age and postnatal maturation time, except for TLR‑4 and TLR‑7. mRNA expression levels of TLR components were significantly increased in the lungs at 45 and 52 dGA, compared with later developmental stages. These results suggest that TLR expression in the guinea pig lung is developmentally regulated, enhancing the understanding of lung biology in guinea pig models.
Collapse
Affiliation(s)
- Lingjie Ma
- Key Laboratory of The Ministry of Education for The Conservation and Utilization of Special Biological Resources of Western China, Ningxia University, Yinchuan, Ningxia 750021, P.R. China
| | - Jiali Yang
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Li Yang
- The Center of Experimental Animals, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Juan Shi
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jing Xue
- Key Laboratory of The Ministry of Education for The Conservation and Utilization of Special Biological Resources of Western China, Ningxia University, Yinchuan, Ningxia 750021, P.R. China
| | - Yong Li
- Key Laboratory of The Ministry of Education for The Conservation and Utilization of Special Biological Resources of Western China, Ningxia University, Yinchuan, Ningxia 750021, P.R. China
| | - Xiaoming Liu
- Key Laboratory of The Ministry of Education for The Conservation and Utilization of Special Biological Resources of Western China, Ningxia University, Yinchuan, Ningxia 750021, P.R. China
| |
Collapse
|
13
|
Campos-Neto A. Mycobacterium tuberculosis strain 18b, a useful non-virulent streptomycin dependent mutant to study latent tuberculosis as well as for in vivo and in vitro testing of anti-tuberculosis drugs. Tuberculosis (Edinb) 2016; 99:54-55. [DOI: 10.1016/j.tube.2016.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/11/2016] [Indexed: 10/21/2022]
|
14
|
Benjak A, Uplekar S, Zhang M, Piton J, Cole ST, Sala C. Genomic and transcriptomic analysis of the streptomycin-dependent Mycobacterium tuberculosis strain 18b. BMC Genomics 2016; 17:190. [PMID: 26944551 PMCID: PMC4779234 DOI: 10.1186/s12864-016-2528-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/24/2016] [Indexed: 12/05/2022] Open
Abstract
Background The ability of Mycobacterium tuberculosis to establish a latent infection (LTBI) in humans confounds the treatment of tuberculosis. Consequently, there is a need to discover new therapeutic agents that can kill M. tuberculosis both during active disease and LTBI. The streptomycin-dependent strain of M. tuberculosis, 18b, provides a useful tool for this purpose since upon removal of streptomycin (STR) it enters a non-replicating state that mimics latency both in vitro and in animal models. Results The 4.41 Mb genome sequence of M. tuberculosis 18b was determined and this revealed the strain to belong to clade 3 of the ancient ancestral lineage of the Beijing family. STR-dependence was attributable to insertion of a single cytosine in the 530 loop of the 16S rRNA and to a single amino acid insertion in the N-terminal domain of initiation factor 3. RNA-seq was used to understand the genetic programme activated upon STR-withdrawal and hence to gain insight into LTBI. This revealed reconfiguration of gene expression and metabolic pathways showing strong similarities between non-replicating 18b and M. tuberculosis residing within macrophages, and with the core stationary phase and microaerophilic responses. Conclusion The findings of this investigation confirm the validity of 18b as a model for LTBI, and provide insight into both the evolution of tubercle bacilli and the functioning of the ribosome. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2528-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrej Benjak
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.
| | - Swapna Uplekar
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland. .,Current addresses: Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| | - Ming Zhang
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland. .,Current addresses: Department of Biochemistry, University of Lausanne, Quartier UNIL-Epalinges, Ch. des Boveresses 155, CH-1066, Epalinges, Switzerland.
| | - Jérémie Piton
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.
| | - Stewart T Cole
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.
| | - Claudia Sala
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.
| |
Collapse
|
15
|
Zhan L, Tang J, Lin S, Xu Y, Xu Y, Qin C. Prophylactic Use of Ganoderma lucidum Extract May Inhibit Mycobacterium tuberculosis Replication in a New Mouse Model of Spontaneous Latent Tuberculosis Infection. Front Microbiol 2016; 6:1490. [PMID: 26779146 PMCID: PMC4705449 DOI: 10.3389/fmicb.2015.01490] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 12/10/2015] [Indexed: 11/30/2022] Open
Abstract
A mouse model of spontaneous latent tuberculosis infection (LTBI) that mimics LTBI in humans is valuable for drug/vaccine development and the study of tuberculosis. However, most LTBI mouse models require interventions, and a spontaneous LTBI mouse model with a low bacterial load is difficult to establish. In this study, mice were IV-inoculated with 100 CFU Mycobacterium tuberculosis H37Rv, and a persistent LTBI was established with low bacterial loads (0.5~1.5log10 CFU in the lung; < 4log10 CFU in the spleen). Histopathological changes in the lung and spleen were mild during the first 20 weeks post-inoculation. The model was used to demonstrate the comparative effects of prophylactic and therapeutic administration of Ganoderma lucidum extract (spores and spores lipid) in preventing H37Rv replication in both lung and spleen. H37Rv was inhibited with prophylactic use of G. lucidum extract relative to that of the untreated control and therapy groups, and observed in the spleen and lung as early as post-inoculation week 3 and week 5 respectively. H37Rv infection in the therapy group was comparable to that of the untreated control mice. No significant mitigation of pathological changes was observed in either the prophylactic or therapeutic group. Our results suggest that this new LTBI mouse model is an efficient tool of testing anti-tuberculosis drug, the use of G. lucidum extract prior to M. tuberculosis infection may protect the host against bacterial replication to some extent.
Collapse
Affiliation(s)
- Lingjun Zhan
- Key Laboratory of Human Diseases and Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Peking Union Medical College, Chinese Academy of Medical Sciences and Comparative Medicine Center Beijing, China
| | - Jun Tang
- Key Laboratory of Human Diseases and Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Peking Union Medical College, Chinese Academy of Medical Sciences and Comparative Medicine Center Beijing, China
| | - Shuzhu Lin
- Key Laboratory of Human Diseases and Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Peking Union Medical College, Chinese Academy of Medical Sciences and Comparative Medicine Center Beijing, China
| | - Yanfeng Xu
- Key Laboratory of Human Diseases and Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Peking Union Medical College, Chinese Academy of Medical Sciences and Comparative Medicine Center Beijing, China
| | - Yuhuan Xu
- Key Laboratory of Human Diseases and Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Peking Union Medical College, Chinese Academy of Medical Sciences and Comparative Medicine Center Beijing, China
| | - Chuan Qin
- Key Laboratory of Human Diseases and Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Peking Union Medical College, Chinese Academy of Medical Sciences and Comparative Medicine Center Beijing, China
| |
Collapse
|
16
|
Braian C, Svensson M, Brighenti S, Lerm M, Parasa VR. A 3D Human Lung Tissue Model for Functional Studies on Mycobacterium tuberculosis Infection. J Vis Exp 2015. [PMID: 26485646 PMCID: PMC4692636 DOI: 10.3791/53084] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Tuberculosis (TB) still holds a major threat to the health of people worldwide, and there is a need for cost-efficient but reliable models to help us understand the disease mechanisms and advance the discoveries of new treatment options. In vitro cell cultures of monolayers or co-cultures lack the three-dimensional (3D) environment and tissue responses. Herein, we describe an innovative in vitro model of a human lung tissue, which holds promise to be an effective tool for studying the complex events that occur during infection with Mycobacterium tuberculosis (M. tuberculosis). The 3D tissue model consists of tissue-specific epithelial cells and fibroblasts, which are cultured in a matrix of collagen on top of a porous membrane. Upon air exposure, the epithelial cells stratify and secrete mucus at the apical side. By introducing human primary macrophages infected with M. tuberculosis to the tissue model, we have shown that immune cells migrate into the infected-tissue and form early stages of TB granuloma. These structures recapitulate the distinct feature of human TB, the granuloma, which is fundamentally different or not commonly observed in widely used experimental animal models. This organotypic culture method enables the 3D visualization and robust quantitative analysis that provides pivotal information on spatial and temporal features of host cell-pathogen interactions. Taken together, the lung tissue model provides a physiologically relevant tissue micro-environment for studies on TB. Thus, the lung tissue model has potential implications for both basic mechanistic and applied studies. Importantly, the model allows addition or manipulation of individual cell types, which thereby widens its use for modelling a variety of infectious diseases that affect the lungs.
Collapse
Affiliation(s)
- Clara Braian
- Department of Clinical and Experimental Medicine, Linköping University
| | | | | | - Maria Lerm
- Department of Clinical and Experimental Medicine, Linköping University;
| | - Venkata R Parasa
- Department of Clinical and Experimental Medicine, Linköping University; Department of Medicine, Karolinska Institute
| |
Collapse
|
17
|
Wali S, Gupta R, Veselenak RL, Li Y, Yu JJ, Murthy AK, Cap AP, Guentzel MN, Chambers JP, Zhong G, Rank RG, Pyles RB, Arulanandam BP. Use of a Guinea pig-specific transcriptome array for evaluation of protective immunity against genital chlamydial infection following intranasal vaccination in Guinea pigs. PLoS One 2014; 9:e114261. [PMID: 25502875 PMCID: PMC4263467 DOI: 10.1371/journal.pone.0114261] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 11/05/2014] [Indexed: 12/21/2022] Open
Abstract
Guinea pigs have been used as a second animal model to validate putative anti-chlamydial vaccine candidates tested in mice. However, the lack of guinea pig-specific reagents has limited the utility of this animal model in Chlamydia sp. vaccine studies. Using a novel guinea pig-specific transcriptome array, we determined correlates of protection in guinea pigs vaccinated with Chlamydia caviae (C. caviae) via the intranasal route, previously reported by us and others to provide robust antigen specific immunity against subsequent intravaginal challenge. C. caviae vaccinated guinea pigs resolved genital infection by day 3 post challenge. In contrast, mock vaccinated animals continued to shed viable Chlamydia up to day 18 post challenge. Importantly, at day 80 post challenge, vaccinated guinea pigs experienced significantly reduced genital pathology - a sequelae of genital chlamydial infections, in comparison to mock vaccinated guinea pigs. Sera from vaccinated guinea pigs displayed antigen specific IgG responses and increased IgG1 and IgG2 titers capable of neutralizing GPIC in vitro. Th1-cellular/inflammatory immune genes and Th2-humoral associated genes were also found to be elevated in vaccinated guinea pigs at day 3 post-challenge and correlated with early clearance of the bacterium. Overall, this study provides the first evidence of guinea pig-specific genes involved in anti-chlamydial vaccination and illustrates the enhancement of the utility of this animal model in chlamydial pathogenesis.
Collapse
Affiliation(s)
- Shradha Wali
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United Stats of America
| | - Rishein Gupta
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United Stats of America
| | - Ronald L. Veselenak
- Departments of Pediatrics and Microbiology & Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555, United States of America
| | - Yansong Li
- US Army Institute of Surgical Research, 3650 Chambers Pass, BHT2, Building 3610/Room224-1, Fort Sam Houston, Texas 78234, United States of America
| | - Jieh-Juen Yu
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United Stats of America
| | - Ashlesh K. Murthy
- Department of Pathology, Midwestern University, Downer's Grove, Illinois, 60148, United States of America
| | - Andrew P. Cap
- US Army Institute of Surgical Research, 3650 Chambers Pass, BHT2, Building 3610/Room224-1, Fort Sam Houston, Texas 78234, United States of America
| | - M. Neal Guentzel
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United Stats of America
| | - James P. Chambers
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United Stats of America
| | - Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, 7702 Floyd Curl Drive, San Antonio, Texas 78229, United States of America
| | - Roger G. Rank
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute, Little Rock, Arkansas 72202, United States of America
| | - Richard B. Pyles
- Departments of Pediatrics and Microbiology & Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555, United States of America
| | - Bernard P. Arulanandam
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United Stats of America
- * E-mail:
| |
Collapse
|
18
|
Parasa VR, Rahman MJ, Ngyuen Hoang AT, Svensson M, Brighenti S, Lerm M. Modeling Mycobacterium tuberculosis early granuloma formation in experimental human lung tissue. Dis Model Mech 2013; 7:281-8. [PMID: 24203885 PMCID: PMC3917249 DOI: 10.1242/dmm.013854] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The widely used animal models for tuberculosis (TB) display fundamental differences from human TB. Therefore, a validated model that recapitulates human lung TB is attractive for TB research. Here, we describe a unique method for establishment of TB infection in an experimental human lung tissue model. The model is based on cell lines derived from human lungs and primary macrophages from peripheral blood, and displays characteristics of human lung tissue, including evenly integrated macrophages throughout the epithelium, production of extracellular matrix, stratified epithelia and mucus secretion. Establishment of experimental infection in the model tissue with Mycobacterium tuberculosis, the bacterium that causes TB, resulted in clustering of macrophages at the site of infection, reminiscent of early TB granuloma formation. We quantitated the extent of granuloma formation induced by different strains of mycobacteria and validated our model against findings in other TB models. We found that early granuloma formation is dependent on ESAT-6, which is secreted via the type VII secretion machinery of virulent mycobacteria. Our model, which can facilitate the discovery of the interactions between mycobacteria and host cells in a physiological environment, is the first lung tissue model described for TB.
Collapse
Affiliation(s)
- Venkata Ramanarao Parasa
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Stockholm 14186, Sweden
| | | | | | | | | | | |
Collapse
|
19
|
Das B, Kashino SS, Pulu I, Kalita D, Swami V, Yeger H, Felsher DW, Campos-Neto A. CD271(+) bone marrow mesenchymal stem cells may provide a niche for dormant Mycobacterium tuberculosis. Sci Transl Med 2013; 5:170ra13. [PMID: 23363977 PMCID: PMC3616630 DOI: 10.1126/scitranslmed.3004912] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mycobacterium tuberculosis (Mtb) can persist in hostile intracellular microenvironments evading immune cells and drug treatment. However, the protective cellular niches where Mtb persists remain unclear. We report that Mtb may maintain long-term intracellular viability in a human bone marrow (BM)-derived CD271(+)/CD45(-) mesenchymal stem cell (BM-MSC) population in vitro. We also report that Mtb resides in an equivalent population of BM-MSCs in a mouse model of dormant tuberculosis infection. Viable Mtb was detected in CD271(+)/CD45(-) BM-MSCs isolated from individuals who had successfully completed months of anti-Mtb drug treatment. These results suggest that CD271(+) BM-MSCs may provide a long-term protective intracellular niche in the host in which dormant Mtb can reside.
Collapse
Affiliation(s)
- Bikul Das
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Suely S. Kashino
- Global Infectious Disease Research Center, Forsyth Institute, Cambridge, MA, USA
| | - Ista Pulu
- Research Institute of World’s Ancient Traditions, Cultures and Heritages (RIWATCH), Roing, Arunachal Pradesh, India
| | - Deepjyoti Kalita
- KaviKrishna Laboratory, Guwahati Biotech Park, Technology Complex, Indian Institute of Technology, Guwahati, India
- Department of Microbiology, Gauhati Medical College, Guwahati, India
| | - Vijay Swami
- Research Institute of World’s Ancient Traditions, Cultures and Heritages (RIWATCH), Roing, Arunachal Pradesh, India
| | - Herman Yeger
- Department of Pediatric Laboratory Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Dean W. Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Antonio Campos-Neto
- Global Infectious Disease Research Center, Forsyth Institute, Cambridge, MA, USA
| |
Collapse
|
20
|
Cayabyab MJ, Macovei L, Campos-Neto A. Current and novel approaches to vaccine development against tuberculosis. Front Cell Infect Microbiol 2012; 2:154. [PMID: 23230563 PMCID: PMC3515764 DOI: 10.3389/fcimb.2012.00154] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 11/20/2012] [Indexed: 11/29/2022] Open
Abstract
Antibiotics and vaccines are the two most successful medical countermeasures that humans have created against a number of pathogens. However a select few e.g., Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB) have evaded eradication by vaccines and therapeutic approaches. TB is a global public health problem that kills 1.4 million people per year. The past decade has seen significant progress in developing new vaccine candidates, but the most fundamental questions in understanding disease progression and protective host responses that are responsible for controlling Mtb infection still remain poorly resolved. Current TB treatment requires intense chemotherapy with several antimicrobials, while the only approved vaccine is the classical viable whole-cell based Bacille-Calmette-Guerin (BCG) that protects children from severe forms of TB, but fails to protect adults. Taken together, there is a growing need to conduct basic and applied research to develop novel vaccine strategies against TB. This review is focused on the discussion surrounding current strategies and innovations being explored to discover new protective antigens, adjuvants, and delivery systems in the hopes of creating an efficacious TB vaccine.
Collapse
Affiliation(s)
- Mark J Cayabyab
- Forsyth Institute Cambridge, MA, USA ; Harvard School of Dental Medicine Boston, MA, USA
| | | | | |
Collapse
|
21
|
Patel K, Jhamb SS, Singh PP. Models of latent tuberculosis: their salient features, limitations, and development. J Lab Physicians 2012; 3:75-9. [PMID: 22219558 PMCID: PMC3249721 DOI: 10.4103/0974-2727.86837] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Latent tuberculosis is a subclinical condition caused by Mycobacterium tuberculosis, which affects about one-third of the population across the world. To abridge the chemotherapy of tuberculosis, it is necessary to have active drugs against latent form of M. tuberculosis. Therefore, it is imperative to devise in vitro and models of latent tuberculosis to explore potential drugs. In vitro models such as hypoxia, nutrient starvation, and multiple stresses are based on adverse conditions encountered by bacilli in granuloma. Bacilli experience oxygen depletion condition in hypoxia model, whereas the nutrient starvation model is based on deprivation of total nutrients from a culture medium. In the multiple stress model dormancy is induced by more than one type of stress. In silico mathematical models have also been developed to predict the interactions of bacilli with the host immune system and to propose structures for potential anti tuberculosis compounds. Besides these in vitro and in silico models, there are a number of in vivo animal models like mouse, guinea pig, rabbit, etc. Although they simulate human latent tuberculosis up to a certain extent but do not truly replicate human infection. All these models have their inherent merits and demerits. However, there is no perfect model for latent tuberculosis. Therefore, it is imperative to upgrade and refine existing models or develop a new model. However, battery of models will always be a better alternative to any single model as they will complement each other by overcoming their limitations.
Collapse
Affiliation(s)
- Kamlesh Patel
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | | | | |
Collapse
|
22
|
Parikka M, Hammarén MM, Harjula SKE, Halfpenny NJA, Oksanen KE, Lahtinen MJ, Pajula ET, Iivanainen A, Pesu M, Rämet M. Mycobacterium marinum causes a latent infection that can be reactivated by gamma irradiation in adult zebrafish. PLoS Pathog 2012; 8:e1002944. [PMID: 23028333 PMCID: PMC3459992 DOI: 10.1371/journal.ppat.1002944] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 08/18/2012] [Indexed: 01/04/2023] Open
Abstract
The mechanisms leading to latency and reactivation of human tuberculosis are still unclear, mainly due to the lack of standardized animal models for latent mycobacterial infection. In this longitudinal study of the progression of a mycobacterial disease in adult zebrafish, we show that an experimental intraperitoneal infection with a low dose (≈ 35 bacteria) of Mycobacterium marinum, results in the development of a latent disease in most individuals. The infection is characterized by limited mortality (25%), stable bacterial loads 4 weeks following infection and constant numbers of highly organized granulomas in few target organs. The majority of bacteria are dormant during a latent mycobacterial infection in zebrafish, and can be activated by resuscitation promoting factor ex vivo. In 5-10% of tuberculosis cases in humans, the disease is reactivated usually as a consequence of immune suppression. In our model, we are able to show that reactivation can be efficiently induced in infected zebrafish by γ-irradiation that transiently depletes granulo/monocyte and lymphocyte pools, as determined by flow cytometry. This immunosuppression causes reactivation of the dormant mycobacterial population and a rapid outgrowth of bacteria, leading to 88% mortality in four weeks. In this study, the adult zebrafish presents itself as a unique non-mammalian vertebrate model for studying the development of latency, regulation of mycobacterial dormancy, as well as reactivation of latent or subclinical tuberculosis. The possibilities for screening for host and pathogen factors affecting the disease progression, and identifying novel therapeutic agents and vaccine targets make this established model especially attractive.
Collapse
|
23
|
Li Y, Wang J, He HY, Ma LJ, Zeng J, Deng GC, Liu X, Engelhardt JF, Wang Y. Immunohistochemical demonstration of airway epithelial cell markers of guinea pig. Tissue Cell 2011; 43:283-90. [PMID: 21705035 DOI: 10.1016/j.tice.2011.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 05/15/2011] [Accepted: 05/24/2011] [Indexed: 11/25/2022]
Abstract
The guinea pig (Cavea porcellus) is a mammalian non-rodent species in the Caviidae family. The sensitivity of the respiratory system and the susceptibility to infectious diseases allows the guinea pig to be a useful model for both infectious and non-infectious lung diseases such as asthma and tuberculosis. In this report, we demonstrated for the first time, the major cell types and composition in the guinea pig airway epithelium, using cell type-specific markers by immunohistochemical staining using the commercial available immunological reagents that cross-react with guinea pig. Our results revealed the availability of antibodies cross-reacting with airway epithelial cell types of basal, non-ciliated columnar, ciliated, Clara, goblet and alveolar type II cells, as well as those cells expressing Mucin 5AC, Mucin 2, Aquaporin 4 and Calcitonin Gene Related Peptide. The distribution of these various cell types were quantified in the guinea pig airway by immunohistochemical staining and were comparable with morphometric studies using an electron microscopy assay. Moreover, this study also demonstrated that goblet cells are the main secretory cell type in the guinea pig's airway, distinguishing this species from rats and mice. These results provide useful information for the understanding of airway epithelial cell biology and mechanisms of epithelial-immune integration in guinea pig models.
Collapse
Affiliation(s)
- Yong Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources of Western China, Yinchuan, Ningxia 750021, China
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Mycobacterium tuberculosis, which causes tuberculosis, remains a major human public health threat. This is largely due to a sizeable reservoir of latently infected individuals, who may relapse into active disease decades after first acquiring the infection. Furthermore, patients have a very slow response to treatment of active disease. Latency and antibiotic tolerance are commonly taken as a proxy for dormancy, a stable nonreplicative state. However, latency is a clinical term that is solely defined by a lack of disease indicators. The actual state of the bacterium in human latency is not well understood. Here we evaluate the results of several in vitro models of dormancy and consider the applicability of various animal models for studying aspects of human latency and resistance to killing by antibiotics. Furthermore, we propose a model for the initiation of dormancy and resuscitation during infection.
Collapse
Affiliation(s)
- Michael C Chao
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
25
|
Simple model for testing drugs against nonreplicating Mycobacterium tuberculosis. Antimicrob Agents Chemother 2010; 54:4150-8. [PMID: 20679505 DOI: 10.1128/aac.00821-10] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nonreplicating or dormant cells of Mycobacterium tuberculosis constitute a challenge to tuberculosis (TB) therapy because of their tolerance or phenotypic resistance to most drugs. Here, we propose a simple model for testing drugs against nongrowing cells that exploits the 18b strain of M. tuberculosis, a streptomycin (STR)-dependent mutant. Optimal conditions were established that allowed 18b cells to replicate in the presence of STR and to survive, but not multiply, following withdrawal of STR. In the presence of the antibiotic, M. tuberculosis 18b was susceptible to the currently approved TB drugs, isoniazid (INH) and rifampin (RIF), and to the experimental drugs TMC207, PA-824, meropenem (MER), benzothiazinone (BTZ), and moxifloxacin (MOXI). After STR depletion, the strain displayed greatly reduced susceptibility to the cell wall inhibitors INH and BTZ but showed increased susceptibility to RIF and PA-824, while MOXI and MER appeared equipotent under both conditions. The same potency ranking was found against nonreplicating M. tuberculosis 18b after in vivo treatment of chronically infected mice with five of these drugs. Despite the growth arrest, strain 18b retains significant metabolic activity in vitro, remaining positive in the resazurin reduction assay. Upon adaption to a 96-well format, this assay was shown to be suitable for high-throughput screening with strain 18b to find new inhibitors of dormant M. tuberculosis.
Collapse
|
26
|
Sugawara I, Udagawa T, Aoki T, Mizuno S. Establishment of a guinea pig model of latent tuberculosis with GFP-introduced Mycobacterium tuberculosis. TOHOKU J EXP MED 2010; 219:257-62. [PMID: 19851055 DOI: 10.1620/tjem.219.257] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
There exists latent tuberculosis, in which small numbers of tubercle bacilli remain viable in the host without visible granulomatous lesions. As few data exist on the mechanisms of latent tuberculosis, it is important to examine latent tuberculosis in terms of pathogenesis and efficacy of chemotherapy. As a first step, we used green fluorescent protein (GFP)-introduced H37Rv Mycobacterium tuberculosis to establish latent tuberculosis in the guinea pig that provides one of the best animal models of tuberculosis. We inoculated the guinea pigs subcutaneously with 100 or 1,000 colony-forming unit (CFU) of tubercle bacilli. During the 300-day follow-up period after infection, there were no clinical signs of disease, suggesting a lack of visible granulomatous lesions. In fact, upon necropsy, no macroscopic tuberculous lesions were recognized, but histopathological examination of the lung, spleen and liver revealed microgranulomas consisting of epithelioid macrophages and lymphocytes without central necrosis. Importantly, photon imaging visualized granulomatous lesions corresponding to these histologically apparent microgranulomas. Tuberculin skin testing of infected guinea pigs showed strong positivity (> or = 10 mm induration) until the end of the experiments. Real-time PCR analysis showed a significant increase in the expression levels of interferon-gamma, tumor necrosis factor-alpha, interleukin-12, and inducible nitric oxide synthase mRNAs in infected lung tissues after 300 days (P < 0.01). As human samples are hardly available to study latent tuberculosis, our guinea pig model would be useful for examining the pathogenesis and molecular mechanisms of latent tuberculosis as well as for monitoring the results of chemotherapy with green fluorescence emission of tubercle bacilli.
Collapse
Affiliation(s)
- Isamu Sugawara
- The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan.
| | | | | | | |
Collapse
|
27
|
Apt A, Kramnik I. Man and mouse TB: contradictions and solutions. Tuberculosis (Edinb) 2009; 89:195-8. [PMID: 19345146 PMCID: PMC2705810 DOI: 10.1016/j.tube.2009.02.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 02/27/2009] [Indexed: 10/20/2022]
|