1
|
Sadeghi MH, Radmehr S, Mohagheghzadeh N, Fathi J, Malekzadegan Y, Moghadam HZ. Innovative electrochemical biosensors for tuberculosis detection. Clin Chim Acta 2025; 574:120327. [PMID: 40286897 DOI: 10.1016/j.cca.2025.120327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Tuberculosis (TB) continues to pose a significant global health threat, highlighting the urgent need for the development of rapid, precise, and accessible diagnostic tools to effectively manage its transmission. Conventional diagnostic techniques, such as sputum microscopy and culture-based assays, face several drawbacks, including lengthy processing times, limited sensitivity, and the requirement for specialized laboratory facilities. In this landscape, electrochemical biosensors have emerged as promising alternatives, offering improved sensitivity, specificity, and rapid detection capabilities. This review presents a thorough overview of recent advancements in the development and application of innovative electrochemical biosensors for TB detection. It explores the integration of nanomaterials such as graphene, gold nanoparticles, and carbon nanotubes, focusing on their contributions to enhanced sensor performance in terms of signal amplification and biorecognition efficacy. By synthesizing current research and technological developments, this review emphasizes the considerable potential of electrochemical biosensors to transform TB diagnostics, ultimately assisting in better disease management and control strategies worldwide.
Collapse
Affiliation(s)
- Mohammad Hassan Sadeghi
- Medical Student, School of Medicine,Zahedan University of Medical Science, Sistanbaluchestan, Iran
| | - Safa Radmehr
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Neda Mohagheghzadeh
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Iran
| | - Javad Fathi
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Iran
| | - Yalda Malekzadegan
- Department of Microbiology and Parasitology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Hesam Zendehdel Moghadam
- Research and Technology Department, Iranian Academic Center for Education Culture and Research Kerman Branch, Iran.
| |
Collapse
|
2
|
Kumari S, Martinez-Garcia J, Ehrman RN, Tang W, Miles J, Basak P, Howlett TS, Wijesundara YH, Wang Z, Izzo A, Restrepo B, Lu L, Meloni G, Gassensmith JJ. Testing the Antigenic Potential of Transmembrane Proteins To Develop a Thermostable Tuberculosis MOF-Liposomal Vaccine. ACS Infect Dis 2025; 11:204-215. [PMID: 39663556 DOI: 10.1021/acsinfecdis.4c00771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Tuberculosis is one of the deadliest infectious diseases and continues to be a major health risk in many parts of the world. Even today, the century-old Bacillus Calmette-Guerin (BCG) vaccine is the only formulation on the market and is ineffective for several sections of the global population responsible for transmission. In the search for antigens that can mount a robust immune response, we have reported the recombinant expression and purification of two novel membrane proteins, the Cation transporter protein V (CtpV) and the Mycobacterial copper transporter B (MctB) present on the membrane surface of Mycobacterium tuberculosis. CtpV was tested as an antigen against the plasma of tuberculosis patients and was found to have a unique immune response profile compared with more commonly studied tuberculosis (TB) antigens. CtpV and MctB were reconstituted into proteoliposomes─individually and in combination─to stabilize them in a lipid bilayer and create a nanoparticle vaccine platform. In vivo experiments demonstrated that when delivered with an adjuvant, these antigens generated a robust Th1-biased T-cell response in mice, with the combination of both antigens performing the best and generating a response comparable to BCG. Since tuberculosis vaccines often need to be shipped to areas with fluctuating power supply, we encapsulated the proteoliposomes and the adjuvant in ZIF-8 to create a shelf-stable formulation. Complementary in vivo studies were carried out to confirm that the ZIF-8 coating did not interfere with or compromise the immunogenicity of the antigens.
Collapse
Affiliation(s)
- Sneha Kumari
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Jonathan Martinez-Garcia
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Ryanne Nicole Ehrman
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Wendy Tang
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Joshua Miles
- Department of Immunology, UT Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Priyanka Basak
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Thomas Sinclair Howlett
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Yalini Hansika Wijesundara
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Ziqi Wang
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Angelo Izzo
- Centenary Institute, University of Sydney, Sydney, NSW2050, Australia
| | - Blanca Restrepo
- School of Public Health at Brownsville, University of Texas Health Science Center at Houston, Brownsville, Texas 78520, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas 78539, United States
| | - Lenette Lu
- Department of Immunology, UT Southwestern Medical Center, Dallas, Texas 75390, United States
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas 75390, United States
- Parkland Health & Hospital System, Dallas, Texas 75235, United States
| | - Gabriele Meloni
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Jeremiah J Gassensmith
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
3
|
Hu Z, Xia J, Wu J, Zhao H, Ji P, Gu L, Gu W, Chen Z, Xu J, Huang X, Ma J, Chen A, Li J, Shu T, Fan XY. A multistage Sendai virus vaccine incorporating latency-associated antigens induces protection against acute and latent tuberculosis. Emerg Microbes Infect 2024; 13:2300463. [PMID: 38164736 PMCID: PMC10769537 DOI: 10.1080/22221751.2023.2300463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
One-quarter of the world's population is infected with Mycobacterium tuberculosis (Mtb). After initial exposure, more immune-competent persons develop asymptomatic latent tuberculosis infection (LTBI) but not active diseases, creates an extensive reservoir at risk of developing active tuberculosis. Previously, we constructed a novel recombinant Sendai virus (SeV)-vectored vaccine encoding two dominant antigens of Mtb, which elicited immune protection against acute Mtb infection. In this study, nine Mtb latency-associated antigens were screened as potential supplementary vaccine candidate antigens, and three antigens (Rv2029c, Rv2028c, and Rv3126c) were selected based on their immune-therapeutic effect in mice, and their elevated immune responses in LTBI human populations. Then, a recombinant SeV-vectored vaccine, termed SeV986A, that expresses three latency-associated antigens and Ag85A was constructed. In murine models, the doses, titers, and inoculation sites of SeV986A were optimized, and its immunogenicity in BCG-primed and BCG-naive mice were determined. Enhanced immune protection against the Mtb challenge was shown in both acute-infection and latent-infection murine models. The expression levels of several T-cell exhaustion markers were significantly lower in the SeV986A-vaccinated group, suggesting that the expression of latency-associated antigens inhibited the T-cell exhaustion process in LTBI infection. Hence, the multistage quarter-antigenic SeV986A vaccine holds considerable promise as a novel post-exposure prophylaxis vaccine against tuberculosis.
Collapse
Affiliation(s)
- Zhidong Hu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | - Jingxian Xia
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | - Juan Wu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | - Huimin Zhao
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | - Ping Ji
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | - Ling Gu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | - Wenfei Gu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | - Zhenyan Chen
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | - Jinchuan Xu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | - Xuejiao Huang
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | | | - Anke Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic of China
| | - Jixi Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic of China
| | | | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
4
|
Wu Y, Xiong Y, Zhong Y, Liao J, Wang J. Role of dormancy survival regulator and resuscitation-promoting factors antigens in differentiating between active and latent tuberculosis: a systematic review and meta-analysis. BMC Pulm Med 2024; 24:541. [PMID: 39472851 PMCID: PMC11523848 DOI: 10.1186/s12890-024-03348-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Dormancy survival regulator (DosR) and resuscitation-promoting factor (Rpf) antigens of Mycobacterium tuberculosis are activated during dormant phase of tuberculosis (TB). This study evaluates the differential immunogenicity potentials of DosR and Rpf antigens in individuals with latent tuberculosis infection (LTBI) and active TB patients. METHODS After a literature search in electronic databases, studies were selected by following precise eligibility criteria. Outcomes were synthesized systematically, and meta-analyses were performed to estimate standardized mean differences (SMDs) in interferon-gamma (IFNγ) levels, and IFNγ positive immune cells between individuals with LTBI and active TB patients. RESULTS Twenty-six studies (1278 individuals with LTBI and 1189 active TB patients) were included. DosR antigens Rv0569 (Standardized mean difference; SMD 2.44 [95%CI: 1.21, 3.66]; p < 0.0001), Rv1733c (SMD 0.60 [95%CI: 0.14, 1.07]; p = 0.011), Rv1735c (SMD 1.16 [95%CI: 0.44, 1.88]; p = 0.002), Rv1737c (SMD 1.26 [95%CI: 0.59, 1.92]; p < 0.0001), Rv2029c (SMD 0.89 [95%CI: 0.35, 1.42]; p = 0.002), RV2626c (SMD 1.24 [95%CI: 0.45, 2.02); p = 0.002), and Rv2628 (SMD 0.65 [95%CI: 0.38, 0.91]; p < 0.0001) and Rpf antigens Rv0867c (SMD 1.33 [95%CI: 0.48, 2.18]; p = 0.002), Rv1009 (SMD 0.65 [95%CI: 0.05, 1.25]; p = 0.034), and Rv2450c (SMD 1.54 [95%CI: 0.92, 2.16]; p < 0.0001) elicited higher IFNγ levels in individuals with LTBI in comparison with active TB patients. IFNγ-positive immunoresponsive cells were significantly higher in individuals with LTBI than in active TB patients for antigens Rv1733c (SMD 1.02 [95%CI: 0.15, 1.88]; p = 0.021), Rv2029c (SMD 0.57 [95%CI: 0.05, 1.09]; p = 0.031), and Rv2628 [SMD 0.38 [95%CI: 0.15, 0.61]; p = 0.001). CONCLUSION DosR antigens Rv0569, Rv1733c, Rv1735c, Rv1737c, RV2626c, Rv2628, and Rv2029c, and Rpf antigens Rv0867c, Rv1009, and Rv2450c are found to elicit immune responses differently in individuals with LTBI and active TB patients.
Collapse
Affiliation(s)
- Yu Wu
- Clinical Laboratory and Blood Transfusion Department, No. 908th Hospital of Joint Logistic Support Force, No. 1028 Jinggangshan Avenue, Qingyunpu District, Nanchang, Jiangxi, 330002, China.
| | - Yuanyuan Xiong
- Clinical Laboratory and Blood Transfusion Department, No. 908th Hospital of Joint Logistic Support Force, No. 1028 Jinggangshan Avenue, Qingyunpu District, Nanchang, Jiangxi, 330002, China
| | - Ying Zhong
- Clinical Laboratory and Blood Transfusion Department, No. 908th Hospital of Joint Logistic Support Force, No. 1028 Jinggangshan Avenue, Qingyunpu District, Nanchang, Jiangxi, 330002, China
| | - Juanjuan Liao
- Clinical Laboratory and Blood Transfusion Department, No. 908th Hospital of Joint Logistic Support Force, No. 1028 Jinggangshan Avenue, Qingyunpu District, Nanchang, Jiangxi, 330002, China
| | - Jin Wang
- Clinical Laboratory and Blood Transfusion Department, No. 908th Hospital of Joint Logistic Support Force, No. 1028 Jinggangshan Avenue, Qingyunpu District, Nanchang, Jiangxi, 330002, China
| |
Collapse
|
5
|
Chen Z, Zhang Y, Wu J, Xu J, Hu Z, Fan XY. A multistage protein subunit vaccine as BCG-booster confers protection against Mycobacterium tuberculosis infection in murine models. Int Immunopharmacol 2024; 139:112811. [PMID: 39068754 DOI: 10.1016/j.intimp.2024.112811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
The eradication of tuberculosis remains a global challenge. Despite being the only licensed vaccine, Bacillus Calmette-Guérin (BCG) confers limited protective efficacy in adults and individuals with latent tuberculosis infections (LTBI). There is an urgent need to develop novel vaccines that can enhance the protective effect of BCG. Protein subunit vaccines have garnered significant research interest due to their safety and plasticity. Based on previous studies, we selected three antigens associated with LTBI (Rv2028c, Rv2029c, Rv3126c) and fused them with an immunodominant antigen Ag85A, resulting in the construction of a multistage protein subunit vaccine named A986. We evaluated the protective effect of recombinant protein A986 adjuvanted with MPL/QS21 as a booster vaccine for BCG against Mycobacterium tuberculosis (Mtb) infection in mice. The A986 + MPL/QS21 induced the secretion of antigen-specific Th1 (IL-2+, IFN-γ+ and TNF-α+) and Th17 (IL-17A+) cytokines in CD4+ and CD8+ T cells within the lung and spleen of mice, while also increased the frequency of central memory and effector memory T cells. Additionally, it also induced the enhanced production of IgG antibodies. Compared to BCG alone, A986 + MPL/QS21 boosting significantly augmented the proliferation of antigen-specific multifunctional T cells and effectively reduced bacterial load in infected mice. Taken together, A986 + MPL/QS21 formulation induced robust antigen-specific immune responses and provided enhanced protection against Mtb infection as a booster of BCG vaccine.
Collapse
Affiliation(s)
- Zhenyan Chen
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China; National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital & The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Ying Zhang
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Juan Wu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Jinchuan Xu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Zhidong Hu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China.
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China; National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital & The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China.
| |
Collapse
|
6
|
Chugh S, Bahal RK, Dhiman R, Singh R. Antigen identification strategies and preclinical evaluation models for advancing tuberculosis vaccine development. NPJ Vaccines 2024; 9:57. [PMID: 38461350 PMCID: PMC10924964 DOI: 10.1038/s41541-024-00834-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 02/05/2024] [Indexed: 03/11/2024] Open
Abstract
In its myriad devastating forms, Tuberculosis (TB) has existed for centuries, and humanity is still affected by it. Mycobacterium tuberculosis (M. tuberculosis), the causative agent of TB, was the foremost killer among infectious agents until the COVID-19 pandemic. One of the key healthcare strategies available to reduce the risk of TB is immunization with bacilli Calmette-Guerin (BCG). Although BCG has been widely used to protect against TB, reports show that BCG confers highly variable efficacy (0-80%) against adult pulmonary TB. Unwavering efforts have been made over the past 20 years to develop and evaluate new TB vaccine candidates. The failure of conventional preclinical animal models to fully recapitulate human response to TB, as also seen for the failure of MVA85A in clinical trials, signifies the need to develop better preclinical models for TB vaccine evaluation. In the present review article, we outline various approaches used to identify protective mycobacterial antigens and recent advancements in preclinical models for assessing the efficacy of candidate TB vaccines.
Collapse
Affiliation(s)
- Saurabh Chugh
- Centre for Tuberculosis Research, Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, Faridabad, 121001, Haryana, India
| | - Ritika Kar Bahal
- Marshall Centre, School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Ramandeep Singh
- Centre for Tuberculosis Research, Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, Faridabad, 121001, Haryana, India.
| |
Collapse
|
7
|
Intranasal multivalent adenoviral-vectored vaccine protects against replicating and dormant M.tb in conventional and humanized mice. NPJ Vaccines 2023; 8:25. [PMID: 36823425 PMCID: PMC9948798 DOI: 10.1038/s41541-023-00623-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Viral-vectored vaccines are highly amenable for respiratory mucosal delivery as a means of inducing much-needed mucosal immunity at the point of pathogen entry. Unfortunately, current monovalent viral-vectored tuberculosis (TB) vaccine candidates have failed to demonstrate satisfactory clinical protective efficacy. As such, there is a need to develop next-generation viral-vectored TB vaccine strategies which incorporate both vaccine antigen design and delivery route. In this study, we have developed a trivalent chimpanzee adenoviral-vectored vaccine to provide protective immunity against pulmonary TB through targeting antigens linked to the three different growth phases (acute/chronic/dormancy) of Mycobacterium tuberculosis (M.tb) by expressing an acute replication-associated antigen, Ag85A, a chronically expressed virulence-associated antigen, TB10.4, and a dormancy/resuscitation-associated antigen, RpfB. Single-dose respiratory mucosal immunization with our trivalent vaccine induced robust, sustained tissue-resident multifunctional CD4+ and CD8+ T-cell responses within the lung tissues and airways, which were further quantitatively and qualitatively improved following boosting of subcutaneously BCG-primed hosts. Prophylactic and therapeutic immunization with this multivalent trivalent vaccine in conventional BALB/c mice provided significant protection against not only actively replicating M.tb bacilli but also dormant, non-replicating persisters. Importantly, when used as a booster, it also provided marked protection in the highly susceptible C3HeB/FeJ mice, and a single respiratory mucosal inoculation was capable of significant protection in a humanized mouse model. Our findings indicate the great potential of this next-generation TB vaccine strategy and support its further clinical development for both prophylactic and therapeutic applications.
Collapse
|
8
|
Liang Y, Li X, Yang Y, Xiao L, Liang Y, Mi J, Xue Y, Gong W, Wang L, Wang J, Zhang J, Shi Y, Peng B, Chen X, Zhao W, Wu X. Preventive effects of Mycobacterium tuberculosis DNA vaccines on the mouse model with latent tuberculosis infection. Front Immunol 2023; 14:1110843. [PMID: 36860878 PMCID: PMC9968874 DOI: 10.3389/fimmu.2023.1110843] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
Background About a quarter of the world's population with latent tuberculosis infection (LTBI) are the main source of active tuberculosis. Bacillus Calmette Guerin (BCG) cannot effectively control LTBI individuals from developing diseases. Latency-related antigens can induce T lymphocytes of LTBI individuals to produce higher IFN-γ levels than tuberculosis patients and normal subjects. Herein, we firstly compared the effects of M. tuberculosis (MTB) ag85ab and 7 latent DNA vaccines on clearing latent MTB and preventing its activation in the mouse LTBI model. Methods A mouse LTBI model was established, and then immunized respectively with PBS, pVAX1 vector, Vaccae vaccine, ag85ab DNA and 7 kinds of latent DNAs (including rv1733c, rv2660c, rv1813c, rv2029c, rv2628, rv2659c and rv3407) for three times. The mice with LTBI were injected with hydroprednisone to activate the latent MTB. Then, the mice were sacrificed for the bacterial count, histopathological examination, and immunological evaluation. Results Using chemotherapy made the MTB latent in the infected mice, and then using hormone treatment reactivated the latent MTB, indicating that the mouse LTBI model was successfully established. After the mouse LTBI model was immunized with the vaccines, the lung colony-forming units (CFUs) and lesion degree of mice in all vaccines group were significantly decreased than those in the PBS group and vector group (P<0.0001, P<0.05). These vaccines could induce antigen-specific cellular immune responses. The number of IFN-γ effector T cells spots secreted by spleen lymphocytes in the ag85ab DNA group was significantly increased than those in the control groups (P<0.05). In the splenocyte culture supernatant, IFN-γ and IL-2 levels in the ag85ab, rv2029c, and rv2659c DNA groups significantly increased (P<0.05), and IL-17A levels in ag85ab and rv2659c DNA groups also significantly increased (P<0.05). Compared with the PBS and vector groups, the proportion of CD4+CD25+FOXP3+ regulatory T cells in spleen lymphocytes of ag85ab, rv2660c, rv2029c, and rv3407 DNA groups were significantly reduced (P<0.05). Conclusions MTB ag85ab and 7 kinds of latent DNA vaccines showed immune preventive efficacies on a mouse model of LTBI, especially the rv2659c, and rv1733c DNA. Our findings will provide candidates for the development of new multi-stage vaccines against TB.
Collapse
Affiliation(s)
- Yan Liang
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Xiaoping Li
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China,Department of Respiration, Hengdong People’s Hospital, Hengyang, China
| | - Yourong Yang
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Li Xiao
- Department of Respiration, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Yumei Liang
- Department of Pathology, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Jie Mi
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Yong Xue
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Wenping Gong
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Lan Wang
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Jie Wang
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Junxian Zhang
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Yingchang Shi
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Bizhen Peng
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Xiaoyang Chen
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Weiguo Zhao
- Department of Respiration, the Eighth Medical Center of PLA General Hospital, Beijing, China,*Correspondence: Weiguo Zhao, ; Xueqiong Wu,
| | - Xueqiong Wu
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China,*Correspondence: Weiguo Zhao, ; Xueqiong Wu,
| |
Collapse
|
9
|
Chin KL, Anibarro L, Sarmiento ME, Acosta A. Challenges and the Way forward in Diagnosis and Treatment of Tuberculosis Infection. Trop Med Infect Dis 2023; 8:tropicalmed8020089. [PMID: 36828505 PMCID: PMC9960903 DOI: 10.3390/tropicalmed8020089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 02/03/2023] Open
Abstract
Globally, it is estimated that one-quarter of the world's population is latently infected with Mycobacterium tuberculosis (Mtb), also known as latent tuberculosis infection (LTBI). Recently, this condition has been referred to as tuberculosis infection (TBI), considering the dynamic spectrum of the infection, as 5-10% of the latently infected population will develop active TB (ATB). The chances of TBI development increase due to close contact with index TB patients. The emergence of multidrug-resistant TB (MDR-TB) and the risk of development of latent MDR-TB has further complicated the situation. Detection of TBI is challenging as the infected individual does not present symptoms. Currently, there is no gold standard for TBI diagnosis, and the only screening tests are tuberculin skin test (TST) and interferon gamma release assays (IGRAs). However, these tests have several limitations, including the inability to differentiate between ATB and TBI, false-positive results in BCG-vaccinated individuals (only for TST), false-negative results in children, elderly, and immunocompromised patients, and the inability to predict the progression to ATB, among others. Thus, new host markers and Mtb-specific antigens are being tested to develop new diagnostic methods. Besides screening, TBI therapy is a key intervention for TB control. However, the long-course treatment and associated side effects result in non-adherence to the treatment. Additionally, the latent MDR strains are not susceptible to the current TBI treatments, which add an additional challenge. This review discusses the current situation of TBI, as well as the challenges and efforts involved in its control.
Collapse
Affiliation(s)
- Kai Ling Chin
- Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
- Borneo Medical and Health Research Centre, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
- Correspondence: (K.L.C.); (L.A.); (A.A.)
| | - Luis Anibarro
- Tuberculosis Unit, Infectious Diseases and Internal Medicine Department, Complexo Hospitalario Universitario de Pontevedra, 36071 Pontevedra, Spain
- Immunology Research Group, Galicia Sur Health Research Institute (IIS-GS), 36312 Vigo, Spain
- Correspondence: (K.L.C.); (L.A.); (A.A.)
| | - Maria E. Sarmiento
- School of Health Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Malaysia
| | - Armando Acosta
- School of Health Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Malaysia
- Correspondence: (K.L.C.); (L.A.); (A.A.)
| |
Collapse
|
10
|
Host biomarker-based quantitative rapid tests for detection and treatment monitoring of tuberculosis and COVID-19. iScience 2022; 26:105873. [PMID: 36590898 PMCID: PMC9791715 DOI: 10.1016/j.isci.2022.105873] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/24/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022] Open
Abstract
Diagnostic services for tuberculosis (TB) are not sufficiently accessible in low-resource settings, where most cases occur, which was aggravated by the COVID-19 pandemic. Early diagnosis of pulmonary TB can reduce transmission. Current TB-diagnostics rely on detection of Mycobacterium tuberculosis (Mtb) in sputum requiring costly, time-consuming methods, and trained staff. In this study, quantitative lateral flow (LF) assays were used to measure levels of seven host proteins in sera from pre-COVID-19 TB patients diagnosed in Europe and latently Mtb-infected individuals (LTBI), and from COVID-19 patients and healthy controls. Analysis of host proteins showed significantly lower levels in LTBI versus TB (AUC:0 · 94) and discriminated healthy individuals from COVID-19 patients (0 · 99) and severe COVID-19 from TB. Importantly, these host proteins allowed treatment monitoring of both respiratory diseases. This study demonstrates the potential of non-sputum LF assays as adjunct diagnostics and treatment monitoring for COVID-19 and TB based on quantitative detection of multiple host biomarkers.
Collapse
|
11
|
Weng S, Zhang J, Ma H, Zhou J, Jia L, Wan Y, Cui P, Ruan Q, Shao L, Wu J, Wang H, Zhang W, Xu Y. B21 DNA vaccine expressing ag85b, rv2029c, and rv1738 confers a robust therapeutic effect against latent Mycobacterium tuberculosis infection. Front Immunol 2022; 13:1025931. [PMID: 36569899 PMCID: PMC9768437 DOI: 10.3389/fimmu.2022.1025931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
Latent tuberculosis infection (LTBI) treatment is known to accelerate the decline in TB incidence, especially in high-risk populations. Mycobacterium tuberculosis (M. tb) expression profiles differ at different growth periods, and vaccines protective and therapeutic effects may increase when they include antigenic compositions from different periods. To develop a post-exposure vaccine that targets LTBI, we constructed four therapeutic DNA vaccines (A39, B37, B31, and B21) using different combinations of antigens from the proliferation phase (Ag85A, Ag85B), PE/PPE family (Rv3425), and latent phase (Rv2029c, Rv1813c, Rv1738). We compared the immunogenicity of the four DNA vaccines in C57BL/6j mice. The B21 vaccine stimulated the strongest cellular immune responses, namely Th1/Th17 and CD8+ cytotoxic T lymphocyte responses. It also induced the generation of strengthened effector memory and central memory T cells. In latently infected mice, the B21 vaccine significantly reduced bacterial loads in the spleens and lungs and decreased lung pathology. In conclusion, the B21 DNA vaccine can enhance T cell responses and control the reactivation of LTBI.
Collapse
Affiliation(s)
- Shufeng Weng
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Jinyi Zhang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Huixia Ma
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Jingyu Zhou
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Liqiu Jia
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanmin Wan
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China,Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Peng Cui
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China,Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiaoling Ruan
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Lingyun Shao
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Wu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Honghai Wang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China,*Correspondence: Ying Xu, ; Wenhong Zhang,
| | - Ying Xu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China,Shanghai Huashen Institute of Microbes and Infections, Shanghai, China,*Correspondence: Ying Xu, ; Wenhong Zhang,
| |
Collapse
|
12
|
Bhatt P, Sharma M, Prakash Sharma P, Rathi B, Sharma S. Mycobacterium tuberculosis dormancy regulon proteins Rv2627c and Rv2628 as Toll like receptor agonist and as potential adjuvant. Int Immunopharmacol 2022; 112:109238. [PMID: 36116151 DOI: 10.1016/j.intimp.2022.109238] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022]
Abstract
During latency, DosR proteins of Mycobacterium tuberculosis (M.tb) get activated and help the bacterium to remain dormant. We have shown earlier that 2 such proteins Rv2627c and Rv2628 are immunogenic and induce a TH1 kind of immune response. In this study, through in-vitro experiments we have confirmed that Rv2627c and Rv2628 proteins act as protein Toll-Like Receptor (TLR) agonist-adjuvant. Rv2627c and Rv2628 stimulated THP-1 macrophages showed an increased expression of TLR2, TLR4 and co-stimulatory molecules CD40, CD80, CD86 and antigen presenting molecule HLA-DR. Further studies also found enhanced expression of downstream signaling molecules of TLR activation like MyD88, NF-κB-p65 and pro-inflammatory cytokines. Inhibition studies using TLR blocking antibodies decreased the expression of co-stimulatory molecules, MyD88, NF-κB-p65, and pro-inflammatory cytokines. Rv2627c and Rv2628 stimulation of HEK-TLR2 reporter cell line confirmed the interaction of these proteins with TLR2. Moreover, molecular docking and simulations of Rv2627c and Rv2628 proteins with TLR2 and TLR4 showed stable interactions. The adjuvant activity of Rv2628 was further validated by a protein adjuvanted with pre-clinically validated peptides as multi-epitope vaccine construct which showed good binding with TLR2 and TLR4 and activate dendritic cells and induce sustained pro-inflammatory cytokine response by C-ImmSim analysis. We propose that our vaccine construct will produce a better immune response than BCG and can be taken up as a post-exposure therapeutic subunit vaccine along with standard TB therapy. We also anticipate that our construct can be taken up as a protein adjuvant with other vaccine candidates as these can activate macrophages through TLR signaling.
Collapse
Affiliation(s)
- Parul Bhatt
- DSKC BioDiscovery Lab, Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| | - Monika Sharma
- DSKC BioDiscovery Lab, Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India
| | - Prem Prakash Sharma
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi 110007, India
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi 110007, India
| | - Sadhna Sharma
- DSKC BioDiscovery Lab, Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| |
Collapse
|
13
|
An Immunoinformatic Strategy to Develop New Mycobacterium tuberculosis Multi-epitope Vaccine. Int J Pept Res Ther 2022; 28:99. [PMID: 35573911 PMCID: PMC9086656 DOI: 10.1007/s10989-022-10406-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2022] [Indexed: 11/12/2022]
Abstract
Mycobacterium tuberculosis causes a life-threatening disease known as tuberculosis (TB). In 2021, tuberculosis was the second cause of death after COVID-19 among infectious diseases. Latent life cycle and development of multidrug resistance in one hand and lack of an effective vaccine in another hand have made TB a global health issue. Here, a multi-epitope vaccine have been designed against TB using five new antigenic protein and immunoinformatic tools. To do so, immunodominant MHC-I/MHC-II binding epitopes of Rv2346, Rv2347, Rv3614, Rv3615 and Rv2031 antigenic proteins have been selected using advanced computational procedures. The vaccine was designed by linking ten epitopes from the antigenic proteins and flagellin and TpD as adjuvant. Three-dimensional (3D) structure of the vaccine was modeled, was refined and was evaluated using bioinformatics tools. The 3D structure of the vaccine was docked into the toll-like-receptors (TLR3, 4, 8) to evaluate potential interaction between the vaccine and TLRs. Evaluation of immunological and physicochemical properties of the constructed vaccine have demonstrated the vaccine construct can induce significant humoral and cellular immune responses, the vaccine is non-allergenic and can be recognized by TLR proteins. The immunoinformatic results reported in the present study demonstrates that it is worth following the designed vaccine by experimental investigations.
Collapse
|
14
|
Carpenter SM, Lu LL. Leveraging Antibody, B Cell and Fc Receptor Interactions to Understand Heterogeneous Immune Responses in Tuberculosis. Front Immunol 2022; 13:830482. [PMID: 35371092 PMCID: PMC8968866 DOI: 10.3389/fimmu.2022.830482] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/07/2022] [Indexed: 12/25/2022] Open
Abstract
Despite over a century of research, Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), continues to kill 1.5 million people annually. Though less than 10% of infected individuals develop active disease, the specific host immune responses that lead to Mtb transmission and death, as well as those that are protective, are not yet fully defined. Recent immune correlative studies demonstrate that the spectrum of infection and disease is more heterogenous than has been classically defined. Moreover, emerging translational and animal model data attribute a diverse immune repertoire to TB outcomes. Thus, protective and detrimental immune responses to Mtb likely encompass a framework that is broader than T helper type 1 (Th1) immunity. Antibodies, Fc receptor interactions and B cells are underexplored host responses to Mtb. Poised at the interface of initial bacterial host interactions and in granulomatous lesions, antibodies and Fc receptors expressed on macrophages, neutrophils, dendritic cells, natural killer cells, T and B cells have the potential to influence local and systemic adaptive immune responses. Broadening the paradigm of protective immunity will offer new paths to improve diagnostics and vaccines to reduce the morbidity and mortality of TB.
Collapse
Affiliation(s)
- Stephen M. Carpenter
- Division of Infectious Disease and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Cleveland Medical Center, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Lenette L. Lu
- Division of Geographic Medicine and Infectious Diseases, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, United States
- Parkland Health and Hospital System, Dallas, TX, United States
| |
Collapse
|
15
|
Gong W, Pan C, Cheng P, Wang J, Zhao G, Wu X. Peptide-Based Vaccines for Tuberculosis. Front Immunol 2022; 13:830497. [PMID: 35173740 PMCID: PMC8841753 DOI: 10.3389/fimmu.2022.830497] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis. As a result of the coronavirus disease 2019 (COVID-19) pandemic, the global TB mortality rate in 2020 is rising, making TB prevention and control more challenging. Vaccination has been considered the best approach to reduce the TB burden. Unfortunately, BCG, the only TB vaccine currently approved for use, offers some protection against childhood TB but is less effective in adults. Therefore, it is urgent to develop new TB vaccines that are more effective than BCG. Accumulating data indicated that peptides or epitopes play essential roles in bridging innate and adaptive immunity and triggering adaptive immunity. Furthermore, innovations in bioinformatics, immunoinformatics, synthetic technologies, new materials, and transgenic animal models have put wings on the research of peptide-based vaccines for TB. Hence, this review seeks to give an overview of current tools that can be used to design a peptide-based vaccine, the research status of peptide-based vaccines for TB, protein-based bacterial vaccine delivery systems, and animal models for the peptide-based vaccines. These explorations will provide approaches and strategies for developing safer and more effective peptide-based vaccines and contribute to achieving the WHO's End TB Strategy.
Collapse
Affiliation(s)
- Wenping Gong
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | - Peng Cheng
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
- Hebei North University, Zhangjiakou City, China
| | - Jie Wang
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xueqiong Wu
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
16
|
Kanaparthi KJ, Afroz S, Minhas G, Moitra A, Khan RA, Medikonda J, Naz S, Cholleti SN, Banerjee S, Khan N. Immunogenic profiling of Mycobacterium tuberculosis DosR protein Rv0569 reveals its ability to switch on Th1 based immunity. Immunol Lett 2022; 242:27-36. [PMID: 35007662 DOI: 10.1016/j.imlet.2022.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 12/15/2021] [Accepted: 01/05/2022] [Indexed: 12/20/2022]
Abstract
Mycobacterium tuberculosis (M.tb) is a multifaceted bacterial pathogen known to infect more than 2 billion people globally. However, a majority of the individuals (>90%) show no overt clinical symptoms of active Tuberculosis (TB) and, it is reported that M.tb in these individuals resides in the latent form. Therefore, huge burden of latently infected population poses serious threat to human health. Inconsistent efficacy of BCG vaccine and poor understanding of latency-associated determinants contribute to the failure of combating M.tb. The discovery of DosR as the master regulator of dormancy, opened new avenues to understand the pathophysiology of the bacterium. Though the specific functions of various DosR genes are yet to be discovered, they have been reported as potent T-cell activators and could elicit strong protective immune responses. Rv0569 is a DosR-encoded conserved hypothetical protein overexpressed during dormancy. However, it is not clearly understood how this protein modulates the host immune response. In the present study, we have demonstrated that Rv0569 has a high antigenic index and induces enhanced secretion of Th1 cytokines IL-12p40 and TNF-α as compared to Th2 cytokine IL-10 in macrophages. Mechanistically, Rv0569 induced the transcription of these pro-inflammatory signatures through the activation of NF-κB pathway. Further, immunization of mice with DosR protein Rv0569 switched the immune response towards Th1-biased cytokine pattern, characterized by the enhanced production of IFN-γ, IL-12p40, and TNF-α. Rv0569 augmented the expansion of antigen-specific IFN-γ and IL-2 producing effector CD4+ and CD8+ T-cells which are hallmarks of Th1 biased protective immunity. Additionally, IgG2a/IgG1 and IgG2b/IgG1 ratio in the serum of immunized mice further confirmed the ability of Rv0569 to skew Th1 biased immune response. In conclusion, we emphasize that Rv0569 has the ability to generate signals to switch on Th1-dominated responses and further suggest that it could be a potential vaccine candidate against latent M.tb infection.
Collapse
Affiliation(s)
- Kala Jyothi Kanaparthi
- Department of Biotechnology and Bioinformatics, School of Life-Sciences, University of Hyderabad, Hyderabad-500046, Telangana, India
| | - Sumbul Afroz
- Department of Biotechnology and Bioinformatics, School of Life-Sciences, University of Hyderabad, Hyderabad-500046, Telangana, India
| | - Gillipsie Minhas
- Department of Biotechnology and Bioinformatics, School of Life-Sciences, University of Hyderabad, Hyderabad-500046, Telangana, India
| | - Anurupa Moitra
- Department of Biotechnology and Bioinformatics, School of Life-Sciences, University of Hyderabad, Hyderabad-500046, Telangana, India
| | - Rafiq Ahmad Khan
- Department of Biotechnology and Bioinformatics, School of Life-Sciences, University of Hyderabad, Hyderabad-500046, Telangana, India
| | - Jayashankar Medikonda
- Department of Biochemistry, School of Life-Sciences, University of Hyderabad, Hyderabad-500046, Telangana, India
| | - Saima Naz
- Department of Biochemistry, School of Life-Sciences, University of Hyderabad, Hyderabad-500046, Telangana, India
| | - Sai Nikhith Cholleti
- Department of Biotechnology and Bioinformatics, School of Life-Sciences, University of Hyderabad, Hyderabad-500046, Telangana, India
| | - Sharmistha Banerjee
- Department of Biochemistry, School of Life-Sciences, University of Hyderabad, Hyderabad-500046, Telangana, India
| | - Nooruddin Khan
- Department of Biotechnology and Bioinformatics, School of Life-Sciences, University of Hyderabad, Hyderabad-500046, Telangana, India.; Department of Animal Biology, School of Life-Sciences, University of Hyderabad, Hyderabad-500046, Telangana, India..
| |
Collapse
|
17
|
Zhang L, Ma H, Wan S, Zhang Y, Gao M, Liu X. Mycobacterium tuberculosis latency-associated antigen Rv1733c SLP improves the accuracy of differential diagnosis of active tuberculosis and latent tuberculosis infection. Chin Med J (Engl) 2022; 135:63-69. [PMID: 34802023 PMCID: PMC8850866 DOI: 10.1097/cm9.0000000000001858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Differential diagnosis of active tuberculosis (ATB) and latent tuberculosis infection (LTBI) has been a challenge for clinicians in high TB burden countries. The purpose of this study was to improve the accuracy of differential diagnosis of ATB and LTBI by using fluorescent immunospot (FluoroSpot) assay to detect specific Th1 cell immune responses. The novel mycobacterium tuberculosis (MTB) latency-associated antigens Rv1733c and synthetic long peptides derived from Rv1733c (Rv1733c SLP) were used based on virulence factors early secreting antigen target-6 (ESAT-6) and culture filtrate protein-10 (CFP-10). METHODS Fifty-seven ATB cases, including 20 pathogen-confirmed ATB and 37 clinically diagnosed ATB, and 36 LTBI cases, were enrolled between January and December 2017. FluoroSpot assay was used to detect the interferon γ (IFN-γ) and interleukin 2 (IL-2) secreted by the specific T cells after being stimulated with MTB virulence factors ESAT-6 and CFP-10, MTB latency-associated antigens Rv1733c and Rv1733c SLP. The receiver operating characteristic (ROC) curve was used to define the best cutoff value of latency-associated antigens in the use of differentiating ATB and LTBI. The sensitivity, specificity, predictive value, and likelihood ratio of ESAT-6 and CFP-10-FluoroSpot combined with latency-associated antigen in the differential diagnosis of ATB and LTBI were also calculated. RESULTS Following the stimulation with Rv1733c and Rv1733c SLP, the frequency of single IL-2-secreting T cells stimulated by Rv1733c SLP had the largest area under the ROC curve, which was 0.766. With a cutoff value of 1 (spot-forming cells [SFCs]/2.5 × 105 peripheral blood mononuclear cells) for frequency, the sensitivity and specificity of distinguishing ATB from LTBI were 72.2% and 73.7%, respectively. ESAT-6 and CFP-10-FluoroSpot detected the frequency and proportion of single IFN-γ-secreting T cells; the sensitivity and specificity of distinguishing ATB from LTBI were 82.5% and 66.7%, respectively. Combined with the frequency of single IL-2-secreting T cells stimulated by Rv1733c SLP on the basis of ESAT-6 and CFP-10-FluoroSpot, the sensitivity and specificity increased to 84.2% and 83.3%, respectively. CONCLUSION Rv1733c SLP, combined with ESAT-6 and CFP-10, might be used as a candidate antigen for T cell-based tuberculosis diagnostic tests to differentiate ATB from LTBI.
Collapse
Affiliation(s)
- Lifan Zhang
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Clinical Epidemiology Unit, Peking Union Medical College, International Clinical Epidemiology Network, Beijing 100730, China
- Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Huimin Ma
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Shijun Wan
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yueqiu Zhang
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Mengqiu Gao
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Xiaoqing Liu
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Clinical Epidemiology Unit, Peking Union Medical College, International Clinical Epidemiology Network, Beijing 100730, China
- Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
18
|
Chiwala G, Liu Z, Mugweru JN, Wang B, Khan SA, Bate PNN, Yusuf B, Hameed HMA, Fang C, Tan Y, Guan P, Hu J, Tan S, Liu J, Zhong N, Zhang T. A recombinant selective drug-resistant M. bovis BCG enhances the bactericidal activity of a second-line anti-tuberculosis regimen. Biomed Pharmacother 2021; 142:112047. [PMID: 34426260 DOI: 10.1016/j.biopha.2021.112047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 10/20/2022] Open
Abstract
Drug-resistant tuberculosis (DR-TB) poses a new threat to global health; to improve the treatment outcome, therapeutic vaccines are considered the best chemotherapy adjuvants. Unfortunately, there is no therapeutic vaccine approved against DR-TB. Our study assessed the therapeutic efficacy of a recombinant drug-resistant BCG (RdrBCG) vaccine in DR-TB. We constructed the RdrBCG overexpressing Ag85B and Rv2628 by selecting drug-resistant BCG strains and transformed them with plasmid pEBCG or pIBCG to create RdrBCG-E and RdrBCG-I respectively. Following successful stability testing, we tested the vaccine's safety in severe combined immune deficient (SCID) mice that lack both T and B lymphocytes plus immunoglobulins. Finally, we evaluated the RdrBCG's therapeutic efficacy in BALB/c mice infected with rifampin-resistant M. tuberculosis and treated with a second-line anti-TB regimen. We obtained M. bovis strains which were resistant to several second-line drugs and M. tuberculosis resistant to rifampin. Notably, the exogenously inserted genes were lost in RdrBCG-E but remained stable in the RdrBCG-I both in vitro and in vivo. When administered adjunct to a second-line anti-TB regimen in a murine model of DR-TB, the RdrBCG-I lowered lung M. tuberculosis burden by 1 log10. Furthermore, vaccination with RdrBCG-I adjunct to chemotherapy minimized lung tissue pathology in mice. Most importantly, the RdrBCG-I showed almost the same virulence as its parent BCG Tice strain in SCID mice. Our findings suggested that the RdrBCG-I was stable, safe and effective as a therapeutic vaccine. Hence, the "recombinant" plus "drug-resistant" BCG strategy could be a useful concept for developing therapeutic vaccines against DR-TB.
Collapse
MESH Headings
- Amikacin/pharmacology
- Amikacin/therapeutic use
- Animals
- Antigens, Bacterial/biosynthesis
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Antitubercular Agents/pharmacology
- Antitubercular Agents/therapeutic use
- BCG Vaccine/biosynthesis
- BCG Vaccine/genetics
- BCG Vaccine/immunology
- BCG Vaccine/therapeutic use
- Disease Models, Animal
- Drug Resistance, Bacterial/genetics
- Levofloxacin/pharmacology
- Levofloxacin/therapeutic use
- Mice, Inbred BALB C
- Mice, SCID
- Mycobacterium bovis/chemistry
- Mycobacterium bovis/drug effects
- Mycobacterium bovis/genetics
- Mycobacterium tuberculosis/drug effects
- Mycobacterium tuberculosis/pathogenicity
- Plasmids
- Prothionamide/pharmacology
- Prothionamide/therapeutic use
- Pyrazinamide/pharmacology
- Pyrazinamide/therapeutic use
- Tuberculosis, Pulmonary/drug therapy
- Tuberculosis, Pulmonary/pathology
- Tuberculosis, Pulmonary/prevention & control
- Vaccines, Synthetic/biosynthesis
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/therapeutic use
- Virulence
- Mice
Collapse
Affiliation(s)
- Gift Chiwala
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong-Hong Kong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou 510530, China; Guangzhou National Laboratory, Guangzhou 510320, China
| | - Zhiyong Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong-Hong Kong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou 510530, China
| | - Julius N Mugweru
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Department of Biological Sciences, University of Embu, Embu 60100, Kenya
| | - Bangxing Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Shahzad Akbar Khan
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong-Hong Kong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou 510530, China
| | - Petuel Ndip Ndip Bate
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong-Hong Kong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou 510530, China; Guangzhou National Laboratory, Guangzhou 510320, China
| | - Buhari Yusuf
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong-Hong Kong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou 510530, China; Guangzhou National Laboratory, Guangzhou 510320, China
| | - H M Adnan Hameed
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong-Hong Kong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou 510530, China; Guangzhou National Laboratory, Guangzhou 510320, China
| | - Cuiting Fang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong-Hong Kong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou 510530, China; Guangzhou National Laboratory, Guangzhou 510320, China
| | - Yaoju Tan
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Ping Guan
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Jinxing Hu
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Shouyong Tan
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Jianxiong Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Nanshan Zhong
- Guangdong-Hong Kong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou 510530, China; Guangzhou National Laboratory, Guangzhou 510320, China; State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Tianyu Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong-Hong Kong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou 510530, China; Guangzhou National Laboratory, Guangzhou 510320, China.
| |
Collapse
|
19
|
Shiraz M, Lata S, Kumar P, Shankar UN, Akif M. Immunoinformatics analysis of antigenic epitopes and designing of a multi-epitope peptide vaccine from putative nitro-reductases of Mycobacterium tuberculosis DosR. INFECTION GENETICS AND EVOLUTION 2021; 94:105017. [PMID: 34332157 DOI: 10.1016/j.meegid.2021.105017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/13/2021] [Accepted: 07/22/2021] [Indexed: 10/20/2022]
Abstract
Mycobacterium tuberculosis (Mtb) resides in alveolar macrophages as a non-dividing and dormant state causing latent tuberculosis. Currently, no vaccine is available against the latent tuberculosis. Latent Mtb expresses ~48 genes under the control of DosR regulon. Among these, putative nitroreductases have significantly high expression levels, help Mtb to cope up with nitrogen stresses and possess antigenic properties. In the current study, immunoinformatics methodologies are applied to predict promiscuous antigenic T-cell epitopes from putative nitro-reductases of the DosR regulon. The promiscuous antigenic T-cell epitopes prediction was performed on the basis of their potential to induce an immune response and forming a stable interaction with the HLA alleles. The highest antigenic promiscuous epitopes were assembled for designing an in-silico vaccine construct. A TLR-2 agonist Phenol-soluble modulin alpha 4 was exploited as an adjuvant. Molecular docking and Molecular Dynamics Simulations were used to predict the stability of vaccine construct with the immune receptor. The predicted promiscuous epitopes may be helpful in the construction of a subunit vaccine against latent tuberculosis, which can also be administered along with the BCG to increase its efficacy. Experimental validation is a prerequisite for the in-silico designed vaccine construct against TB infection.
Collapse
Affiliation(s)
- Mohd Shiraz
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Surabhi Lata
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Pankaj Kumar
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Umate Nachiket Shankar
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Mohd Akif
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India.
| |
Collapse
|
20
|
Morgan J, Muskat K, Tippalagama R, Sette A, Burel J, Lindestam Arlehamn CS. Classical CD4 T cells as the cornerstone of antimycobacterial immunity. Immunol Rev 2021; 301:10-29. [PMID: 33751597 PMCID: PMC8252593 DOI: 10.1111/imr.12963] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/11/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022]
Abstract
Tuberculosis is a significant health problem without an effective vaccine to combat it. A thorough understanding of the immune response and correlates of protection is needed to develop a more efficient vaccine. The immune response against Mycobacterium tuberculosis (Mtb) is complex and involves all aspects of the immune system, however, the optimal protective, non‐pathogenic T cell response against Mtb is still elusive. This review will focus on discussing CD4 T cell immunity against mycobacteria and its importance in Mtb infection with a primary focus on human studies. We will in particular discuss the large heterogeneity of immune cell subsets that have been revealed by recent immunological investigations at an unprecedented level of detail. These studies have identified specific classical CD4 T cell subsets important for immune responses against Mtb in various states of infection. We further discuss the functional attributes that have been linked to the various subsets such as upregulation of activation markers and cytokine production. Another important topic to be considered is the antigenic targets of Mtb‐specific immune responses, and how antigen reactivity is influenced by both disease state and environmental exposure(s). These are key points for both vaccines and immune diagnostics development. Ultimately, these factors are holistically considered in the definition and investigations of what are the correlates on protection and resolution of disease.
Collapse
Affiliation(s)
- Jeffrey Morgan
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Kaylin Muskat
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Rashmi Tippalagama
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Julie Burel
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | | |
Collapse
|
21
|
Meier NR, Sutter TM, Jacobsen M, Ottenhoff THM, Vogt JE, Ritz N. Machine Learning Algorithms Evaluate Immune Response to Novel Mycobacterium tuberculosis Antigens for Diagnosis of Tuberculosis. Front Cell Infect Microbiol 2021; 10:594030. [PMID: 33489933 PMCID: PMC7820115 DOI: 10.3389/fcimb.2020.594030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/24/2020] [Indexed: 12/20/2022] Open
Abstract
Rationale Tuberculosis diagnosis in children remains challenging. Microbiological confirmation of tuberculosis disease is often lacking, and standard immunodiagnostic including the tuberculin skin test and interferon-γ release assay for tuberculosis infection has limited sensitivity. Recent research suggests that inclusion of novel Mycobacterium tuberculosis antigens has the potential to improve standard immunodiagnostic tests for tuberculosis. Objective To identify optimal antigen–cytokine combinations using novel Mycobacterium tuberculosis antigens and cytokine read-outs by machine learning algorithms to improve immunodiagnostic assays for tuberculosis. Methods A total of 80 children undergoing investigation of tuberculosis were included (15 confirmed tuberculosis disease, five unconfirmed tuberculosis disease, 28 tuberculosis infection and 32 unlikely tuberculosis). Whole blood was stimulated with 10 novel Mycobacterium tuberculosis antigens and a fusion protein of early secretory antigenic target (ESAT)-6 and culture filtrate protein (CFP) 10. Cytokines were measured using xMAP multiplex assays. Machine learning algorithms defined a discriminative classifier with performance measured using area under the receiver operating characteristics. Measurements and main results We found the following four antigen–cytokine pairs had a higher weight in the discriminative classifier compared to the standard ESAT-6/CFP-10-induced interferon-γ: Rv2346/47c- and Rv3614/15c-induced interferon-gamma inducible protein-10; Rv2031c-induced granulocyte-macrophage colony-stimulating factor and ESAT-6/CFP-10-induced tumor necrosis factor-α. A combination of the 10 best antigen–cytokine pairs resulted in area under the curve of 0.92 ± 0.04. Conclusion We exploited the use of machine learning algorithms as a key tool to evaluate large immunological datasets. This identified several antigen–cytokine pairs with the potential to improve immunodiagnostic tests for tuberculosis in children.
Collapse
Affiliation(s)
- Noëmi Rebecca Meier
- Mycobacterial Research Laboratory, University of Basel Children's Hospital, Basel, Switzerland.,Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Thomas M Sutter
- Department of Computer Science, Medical Data Science, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Marc Jacobsen
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Heinreich Heine University, Düsseldorf, Germany
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Julia E Vogt
- Department of Computer Science, Medical Data Science, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Nicole Ritz
- Mycobacterial Research Laboratory, University of Basel Children's Hospital, Basel, Switzerland.,Faculty of Medicine, University of Basel, Basel, Switzerland.,Pediatric Infectious Diseases and Vaccinology Unit, University of Basel Children's Hospital, Basel, Switzerland.,Department of Pediatrics, Royal Children's Hospital Melbourne, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
22
|
Identification and in silico functional prediction of lineage-specific SNPs distributed in DosR-related proteins and resuscitation-promoting factor proteins of Mycobacterium tuberculosis. Heliyon 2020; 6:e05744. [PMID: 33364506 PMCID: PMC7753917 DOI: 10.1016/j.heliyon.2020.e05744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/21/2020] [Accepted: 12/11/2020] [Indexed: 11/23/2022] Open
Abstract
One-third of the world population is infected by Mycobacterium tuberculosis, which may persist in the latent or dormant state. Bacteria can shift to dormancy when encountering harsh conditions such as low oxygen, nutrient starvation, high acidity and host immune defenses. Genes related to the dormancy survival regulator (DosR) regulon are responsible for the inhibition of aerobic respiration and replication, which is required to enter dormancy. Conversely, resuscitation-promoting factor (rpf) proteins participate in reactivation from dormancy and the development of active tuberculosis (TB). Many DosR regulon and rpf proteins are immunodominant T cell antigens that are highly expressed in latent TB infection. They could serve as TB vaccine candidates and be used for diagnostic development. We explored the genetic polymorphisms of 50 DosR-related genes and 5 rpf genes among 1,170 previously sequenced clinical M. tuberculosis genomes. Forty-three lineage- or sublineage-specific nonsynonymous single nucleotide polymorphisms (nsSNPs) were identified. Ten nsSNPs were specific to all Mtb isolates belonging to lineage 1 (L1). Two common sublineages, the Beijing family (L2.2) and EAI2 (L1.2.1), differed at as many as 26 lineage- or sublineage-specific SNPs. DosR regulon genes related to membrane proteins and the rpf family possessed mean dN/dS ratios greater than one, suggesting that they are under positive selection. Although the T cell epitope regions of DosR-related and rpf antigens were quite conserved, we found that the epitopes in L1 had higher rates of genetic polymorphisms than the other lineages. Some mutations in immunogenic epitopes of the antigens were specific to particular M. tuberculosis lineages. Therefore, the genetic diversity of the DosR regulon and rpf proteins might impact the adaptation of M. tuberculosis to the dormant state and the immunogenicity of latency antigens, which warrants further investigation.
Collapse
|
23
|
Ma A, Wang C, Chang Y, Brennan FH, McDermaid A, Liu B, Zhang C, Popovich PG, Ma Q. IRIS3: integrated cell-type-specific regulon inference server from single-cell RNA-Seq. Nucleic Acids Res 2020; 48:W275-W286. [PMID: 32421805 PMCID: PMC7319566 DOI: 10.1093/nar/gkaa394] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/25/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022] Open
Abstract
A group of genes controlled as a unit, usually by the same repressor or activator gene, is known as a regulon. The ability to identify active regulons within a specific cell type, i.e., cell-type-specific regulons (CTSR), provides an extraordinary opportunity to pinpoint crucial regulators and target genes responsible for complex diseases. However, the identification of CTSRs from single-cell RNA-Seq (scRNA-Seq) data is computationally challenging. We introduce IRIS3, the first-of-its-kind web server for CTSR inference from scRNA-Seq data for human and mouse. IRIS3 is an easy-to-use server empowered by over 20 functionalities to support comprehensive interpretations and graphical visualizations of identified CTSRs. CTSR data can be used to reliably characterize and distinguish the corresponding cell type from others and can be combined with other computational or experimental analyses for biomedical studies. CTSRs can, therefore, aid in the discovery of major regulatory mechanisms and allow reliable constructions of global transcriptional regulation networks encoded in a specific cell type. The broader impact of IRIS3 includes, but is not limited to, investigation of complex diseases hierarchies and heterogeneity, causal gene regulatory network construction, and drug development. IRIS3 is freely accessible from https://bmbl.bmi.osumc.edu/iris3/ with no login requirement.
Collapse
Affiliation(s)
- Anjun Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Cankun Wang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Yuzhou Chang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Faith H Brennan
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Belford Center for Spinal Cord Injury, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Adam McDermaid
- Imagenetics, Sanford Health, Sioux Falls, SD 57104, USA.,Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| | - Bingqiang Liu
- School of Mathematics, Shandong University, Jinan 250100, China
| | - Chi Zhang
- Department of Medical & Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Phillip G Popovich
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Belford Center for Spinal Cord Injury, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
24
|
Carranza C, Pedraza-Sanchez S, de Oyarzabal-Mendez E, Torres M. Diagnosis for Latent Tuberculosis Infection: New Alternatives. Front Immunol 2020; 11:2006. [PMID: 33013856 PMCID: PMC7511583 DOI: 10.3389/fimmu.2020.02006] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/24/2020] [Indexed: 12/29/2022] Open
Abstract
Latent tuberculosis infection (LTBI) is a subclinical mycobacterial infection defined on the basis of cellular immune response to mycobacterial antigens. The tuberculin skin test (TST) and the interferon gamma release assay (IGRA) are currently used to establish the diagnosis of LTB. However, neither TST nor IGRA is useful to discriminate between active and latent tuberculosis. Moreover, these tests cannot be used to predict whether an individual with LTBI will develop active tuberculosis (TB) or whether therapy for LTBI could be effective to decrease the risk of developing active TB. Therefore, in this article, we review current approaches and some efforts to identify an immunological marker that could be useful in distinguishing LTBI from TB and in evaluating the effectiveness of treatment of LTB on the risk of progression to active TB.
Collapse
Affiliation(s)
- Claudia Carranza
- Departamento de Microbiología, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Sigifredo Pedraza-Sanchez
- Unidad de Bioquímica Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, Mexico City, Mexico
| | | | - Martha Torres
- Departamento de Microbiología, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico.,Subdirección de Investigación Biomédica, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| |
Collapse
|
25
|
Keikha M, Eslami M, Yousefi B, Karbalaei M. Overview of multistage subunit tuberculosis vaccines: advantages and challenges. REVIEWS IN MEDICAL MICROBIOLOGY 2020; 31:144-149. [DOI: 10.1097/mrm.0000000000000213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To date, tuberculosis (TB) infection, is the most threatening infectious disease in all humans around the world. Mycobacterium tuberculosis is a facultative intracellular bacterium, possesses an exclusive life-cycle inside the macrophages, as one of the most important cells in the innate immune system. As soon as entrance in the lungs, bacteria actively replicate, but intracellular conditions such as hypoxia and nutrient starvation, lead to low replication of bacteria, or nonreplicating state. While Bacillus Calmette-Guerin vaccine is the most usable vaccine, especially in children and against active form, but this vaccine has no more protection in infected adults to latent forms of disease. Among the new generation of vaccines, fusion multistage subunit vaccines have prodigious effect on immune responses. By virtue of simultaneous presence of both expressed antigens from active and latent forms of TB in the structure of these recombinant subunit vaccines, they can strongly induce immune responses against all stages of the disease. The findings suggest subunit vaccines are the best candidates for immunization against TB, by virtue of their high safety, ease of production, specificity, and utilization of mycobacterial immunodominant antigens. Fusion multistage subunit vaccines, as novel subunit vaccines are the most ideal target for proper prevention against TB infection. Due to simultaneous use of both expressed antigens in active and latent forms of TB, these vaccines are able to induce strong immune responses versus all of TB stages.
Collapse
Affiliation(s)
- Masoud Keikha
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad
| | | | - Bahman Yousefi
- Department of Immunology, Semnan University of Medical Sciences, Semnan
| | - Mohsen Karbalaei
- Department of Microbiology and Virology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| |
Collapse
|
26
|
Gallant J, Mouton J, Ummels R, Ten Hagen-Jongman C, Kriel N, Pain A, Warren RM, Bitter W, Heunis T, Sampson SL. Identification of gene fusion events in Mycobacterium tuberculosis that encode chimeric proteins. NAR Genom Bioinform 2020; 2:lqaa033. [PMID: 33575588 PMCID: PMC7671302 DOI: 10.1093/nargab/lqaa033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/16/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023] Open
Abstract
Mycobacterium tuberculosis is a facultative intracellular pathogen responsible for causing tuberculosis. The harsh environment in which M. tuberculosis survives requires this pathogen to continuously adapt in order to maintain an evolutionary advantage. However, the apparent absence of horizontal gene transfer in M. tuberculosis imposes restrictions in the ways by which evolution can occur. Large-scale changes in the genome can be introduced through genome reduction, recombination events and structural variation. Here, we identify a functional chimeric protein in the ppe38-71 locus, the absence of which is known to have an impact on protein secretion and virulence. To examine whether this approach was used more often by this pathogen, we further develop software that detects potential gene fusion events from multigene deletions using whole genome sequencing data. With this software we could identify a number of other putative gene fusion events within the genomes of M. tuberculosis isolates. We were able to demonstrate the expression of one of these gene fusions at the protein level using mass spectrometry. Therefore, gene fusions may provide an additional means of evolution for M. tuberculosis in its natural environment whereby novel chimeric proteins and functions can arise.
Collapse
Affiliation(s)
- James Gallant
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Science, Faculty of Medicine and Health Science, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa.,Section of Molecular Microbiology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Jomien Mouton
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Science, Faculty of Medicine and Health Science, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa
| | - Roy Ummels
- Medical Microbiology and Infection Control, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Corinne Ten Hagen-Jongman
- Section of Molecular Microbiology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Nastassja Kriel
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Science, Faculty of Medicine and Health Science, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa
| | - Arnab Pain
- Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia.,Global Station for Zoonosis Control, GI-CoRE, Hokkaido University, 001-0020, N20 W10 Kita-ku, Sapporo, Japan
| | - Robin M Warren
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Science, Faculty of Medicine and Health Science, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa
| | - Wilbert Bitter
- Section of Molecular Microbiology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands.,Medical Microbiology and Infection Control, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Tiaan Heunis
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Science, Faculty of Medicine and Health Science, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa.,Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Samantha L Sampson
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Science, Faculty of Medicine and Health Science, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa
| |
Collapse
|
27
|
Gaps in Study Design for Immune Parameter Research for Latent Tuberculosis Infection: A Systematic Review. J Immunol Res 2020; 2020:8074183. [PMID: 32377537 PMCID: PMC7191376 DOI: 10.1155/2020/8074183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 03/16/2020] [Indexed: 01/09/2023] Open
Abstract
Background Immune parameters (IP) have been extensively studied to distinguish between latent tuberculosis (LTBI) and active tuberculosis (TB). Objective To determine the IP associated with LTBI, compared to active TB and individuals not infected by M. tuberculosis published in literature. Methods We conducted a systematic search using Google Scholar and PubMed databases, combining the MeSH terms latent tuberculosis, Mycobacterium tuberculosis, cytokines, and biological markers, with the free terms, biomarkers and cytokines. Spanish, English, and Portuguese articles comparing the concentration of IP associated with LTBI, either in plasma/serum or in vitro, in adults and nonimmunocompromised versus individuals with TB or without M. tuberculosis infection between 2006 July and 2018 July were included. Two blinded reviewers carried out the searches, read the abstracts, and selected the articles for analysis. Participants' information, diagnostic criteria, IP, detection methods, and biases were collected. Results We analyzed 36 articles (of 637 abstracts) with 93 different biomarkers in different samples. We found 24 parameters that were increased only in active TB (TGF-α, CSF3, CSF2, CCL1 [I-309], IL-7, TGF-β1, CCL3 [MIP-1α], sIL-2R, TNF-β, CCL7 [MCP-3], IFN-α, fractalkine, I-TAG, CCL8 [MCP-2], CCL21 [6Ckine], PDGF, IL-22, VEGF-A, LXA4, PGE2, PGF2α, sCD163, sCD14, and 15-Epi-LXA4), five were elevated in LTBI (IL-5, IL-17F, IL-1, CCL20 [MIP-3α], and ICAM-1), and two substances were increased among uninfected individuals (IL-23 and basic FGF). We found high heterogeneity between studies including failure to account for the time/illness of the individuals studied; varied samples and protocols; different clinical classification of TB; different laboratory methods for IP detection, which in turn leads to variable units of measurement and assay sensitivities; and selection bias regarding TST and booster effect. None of the studies adjusted the analysis for the effect of ethnicity. Conclusions It is mandatory to harmonize the study of immune parameters for LTBI diagnosis. This systematic review is registered with PROSPERO CRD42017073289.
Collapse
|
28
|
Shi SD, Hsueh PR, Yang PC, Chou CC. Use of DosR Dormancy Antigens from Mycobacterium tuberculosis for Serodiagnosis of Active and Latent Tuberculosis. ACS Infect Dis 2020; 6:272-280. [PMID: 31815418 DOI: 10.1021/acsinfecdis.9b00329] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
As more than two billion people possibly have a latent tuberculosis (LTB) infection, early LTB diagnosis is crucial for the efficient control and elimination of tuberculosis (TB). The aim of this study is to detect the serum antibody responses to dormancy-related DosR regulon antigens of Mycobacterium tuberculosis for the diagnosis of active and latent TB infections. A membrane array with 25 latency antigens detected by silver-enhanced gold nanoparticles was used to determine the corresponding cognate antibody levels in clinical serum samples from healthy controls, TB patients, and individuals with LTB. The array is sized to fit into a 24-well ELISA plate and follows an ELISA-like experimental procedure without expensive instrumentation. Linear discriminant analysis (LDA) of the resulting antibody profiling data set identified a panel of nine DosR antigens with significant discriminatory capability among different subjects with ≥90% sensitivity, specificity, and overall accuracy. Furthermore, the high predictive performance validated by an independent test sample set reflects the robustness and reliability of the LDA classification model. Our current data demonstrate that the nine DosR antigen combination associated with the proposed membrane array platform is a clinically feasible approach for distinguishing different TB infection statuses. The proposed methodology in this study could be further developed for multiple disease serodiagnoses with high sensitivity and specificity.
Collapse
Affiliation(s)
- Sheng-Dong Shi
- Department of Biomedical Sciences and Institute of Molecular Biology, National Chung Cheng University, No. 168 University Road, Min-Hsiung, Chia-Yi, Taiwan 62102, ROC
- Division of Laboratory Medicine, Chia-Yi and Wanqiao Branch, Taichung Veterans General Hospital, No. 600 Shixian Rd., Chia-Yi City, Taiwan 60090, ROC
| | - Po-Ren Hsueh
- Department of Laboratory Medicine, National Taiwan University College of Medicine, No. 1 Jen Ai Road, Taipei, Taiwan 10051, ROC
| | - Pan-Chyr Yang
- Department of Internal Medicine, National Taiwan University College of Medicine, No. 1 Jen Ai Road, Taipei, Taiwan 10051, ROC
| | - Cheng-Chung Chou
- Department of Biomedical Sciences and Institute of Molecular Biology, National Chung Cheng University, No. 168 University Road, Min-Hsiung, Chia-Yi, Taiwan 62102, ROC
| |
Collapse
|
29
|
Coppola M, Villar-Hernández R, van Meijgaarden KE, Latorre I, Muriel Moreno B, Garcia-Garcia E, Franken KLMC, Prat C, Stojanovic Z, De Souza Galvão ML, Millet JP, Sabriá J, Sánchez-Montalva A, Noguera-Julian A, Geluk A, Domínguez J, Ottenhoff THM. Cell-Mediated Immune Responses to in vivo-Expressed and Stage-Specific Mycobacterium tuberculosis Antigens in Latent and Active Tuberculosis Across Different Age Groups. Front Immunol 2020; 11:103. [PMID: 32117257 PMCID: PMC7026259 DOI: 10.3389/fimmu.2020.00103] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
A quarter of the global human population is estimated to be latently infected by Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). TB remains the global leading cause of death by a single pathogen and ranks among the top-10 causes of overall global mortality. Current immunodiagnostic tests cannot discriminate between latent, active and past TB, nor predict progression of latent infection to active disease. The only registered TB vaccine, Bacillus Calmette-Guérin (BCG), does not adequately prevent pulmonary TB in adolescents and adults, thus permitting continued TB-transmission. Several Mtb proteins, mostly discovered through IFN-γ centered approaches, have been proposed as targets for new TB-diagnostic tests or -vaccines. Recently, however, we identified novel Mtb antigens capable of eliciting multiple cytokines, including antigens that did not induce IFN-γ but several other cytokines. These antigens had been selected based on high Mtb gene-expression in the lung in vivo, and have been termed in vivo expressed (IVE-TB) antigens. Here, we extend and validate our previous findings in an independent Southern European cohort, consisting of adults and adolescents with either LTBI or TB. Our results confirm that responses to IVE-TB antigens, and also DosR-regulon and Rpf stage-specific Mtb antigens are marked by multiple cytokines, including strong responses, such as for TNF-α, in the absence of detectable IFN-γ production. Except for TNF-α, the magnitude of those responses were significantly higher in LTBI subjects. Additional unbiased analyses of high dimensional flow-cytometry data revealed that TNF-α+ cells responding to Mtb antigens comprised 17 highly heterogeneous cell types. Among these 17 TNF-α+ cells clusters identified, those with CD8+TEMRA or CD8+CD4+ phenotypes, defined by the expression of multiple intracellular markers, were the most prominent in adult LTBI, while CD14+ TNF-α+ myeloid-like clusters were mostly abundant in adolescent LTBI. Our findings, although limited to a small cohort, stress the importance of assessing broader immune responses than IFN-γ alone in Mtb antigen discovery as well as the importance of screening individuals of different age groups. In addition, our results provide proof of concept showing how unbiased multidimensional multiparametric cell subset analysis can identify unanticipated blood cell subsets that could play a role in the immune response against Mtb.
Collapse
Affiliation(s)
- Mariateresa Coppola
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Raquel Villar-Hernández
- Institut d'Investigació Germans Trias i Pujol, CIBER Enfermedades Respiratorias, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Irene Latorre
- Institut d'Investigació Germans Trias i Pujol, CIBER Enfermedades Respiratorias, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Beatriz Muriel Moreno
- Institut d'Investigació Germans Trias i Pujol, CIBER Enfermedades Respiratorias, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Esther Garcia-Garcia
- Institut d'Investigació Germans Trias i Pujol, CIBER Enfermedades Respiratorias, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Cristina Prat
- Institut d'Investigació Germans Trias i Pujol, CIBER Enfermedades Respiratorias, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Zoran Stojanovic
- Servei de Neumología Hospital Universitari Germans Trias i Pujol, Institut d'Investigació Germans Trias i Pujol, CIBER Enfermedades Respiratorias, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Joan-Pau Millet
- Serveis Clínics, Unitat Clínica de Tractament Directament Observat de la Tuberculosi, CIBER de Epidemiología y Salud Pública (CIBEREESP), Madrid, Spain
| | - Josefina Sabriá
- Servei de Pneumologia, Hospital Sant Joan Despí Moises Broggi, Sant Joan Despí, Spain
| | - Adrián Sánchez-Montalva
- Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Universitat Autònoma de Barcelona, Barcelona, Spain.,Grupo de Estudio de Micobacterias (GEIM), Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), Madrid, Spain
| | - Antoni Noguera-Julian
- Malalties Infeccioses i Resposta Inflamatòria Sistèmica en Pediatria, Unitat d'Infeccions, Servei de Pediatria, Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Jose Domínguez
- Institut d'Investigació Germans Trias i Pujol, CIBER Enfermedades Respiratorias, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
30
|
Gutti G, Arya K, Singh SK. Latent Tuberculosis Infection (LTBI) and Its Potential Targets: An Investigation into Dormant Phase Pathogens. Mini Rev Med Chem 2019; 19:1627-1642. [PMID: 31241015 DOI: 10.2174/1389557519666190625165512] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/06/2018] [Accepted: 05/28/2018] [Indexed: 11/22/2022]
Abstract
One-third of the world's population harbours the latent tuberculosis infection (LTBI) with a lifetime risk of reactivation. Although, the treatment of LTBI relies significantly on the first-line therapy, identification of novel drug targets and therapies are the emerging focus for researchers across the globe. The current review provides an insight into the infection, diagnostic methods and epigrammatic explanations of potential molecular targets of dormant phase bacilli. This study also includes current preclinical and clinical aspects of tubercular infections and new approaches in antitubercular drug discovery.
Collapse
Affiliation(s)
- Gopichand Gutti
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.) Varanasi-221005 (U.P.), India
| | - Karan Arya
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.) Varanasi-221005 (U.P.), India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.) Varanasi-221005 (U.P.), India
| |
Collapse
|
31
|
Gautam US, Mehra S, Kumari P, Alvarez X, Niu T, Tyagi JS, Kaushal D. Mycobacterium tuberculosis sensor kinase DosS modulates the autophagosome in a DosR-independent manner. Commun Biol 2019; 2:349. [PMID: 31552302 PMCID: PMC6754383 DOI: 10.1038/s42003-019-0594-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 09/03/2019] [Indexed: 01/03/2023] Open
Abstract
Dormancy is a key characteristic of the intracellular life-cycle of Mtb. The importance of sensor kinase DosS in mycobacteria are attributed in part to our current findings that DosS is required for both persistence and full virulence of Mtb. Here we show that DosS is also required for optimal replication in macrophages and involved in the suppression of TNF-α and autophagy pathways. Silencing of these pathways during the infection process restored full virulence in MtbΔdosS mutant. Notably, a mutant of the response regulator DosR did not exhibit the attenuation in macrophages, suggesting that DosS can function independently of DosR. We identified four DosS targets in Mtb genome; Rv0440, Rv2859c, Rv0994, and Rv0260c. These genes encode functions related to hypoxia adaptation, which are not directly controlled by DosR, e.g., protein recycling and chaperoning, biosynthesis of molybdenum cofactor and nitrogen metabolism. Our results strongly suggest a DosR-independent role for DosS in Mtb.
Collapse
Affiliation(s)
- Uma S. Gautam
- Tulane National Primate Research Center, Covington, LA 70433 USA
- Present Address: Duke Human Vaccine Institute, Duke University School of Medicine, 909 S. LaSalle St., Durham, NC 27710 USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, LA 70433 USA
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803 USA
- Center for Experimental Infectious Diseases Research, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803 USA
| | - Priyanka Kumari
- All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Xavier Alvarez
- Tulane National Primate Research Center, Covington, LA 70433 USA
| | - Tianhua Niu
- Department of Biochemistry, Tulane University School of Medicine, New Orleans, 70112 LA USA
| | - Jaya S. Tyagi
- All India Institute of Medical Sciences, New Delhi, 110029 India
- Centre for Bio-design and Diagnostics, Translational Health Science and Technology Institute Faridabad, Haryana, 121001 India
| | - Deepak Kaushal
- Tulane National Primate Research Center, Covington, LA 70433 USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, 70112 LA USA
| |
Collapse
|
32
|
Kasempimolporn S, Areekul P, Thaveekarn W, Sutthisri R, Boonchang S, Sawangvaree A, Sitprija V. Application of transdermal patches with new skin test reagents for detection of latent tuberculosis. J Med Microbiol 2019; 68:1314-1319. [PMID: 31274404 DOI: 10.1099/jmm.0.001037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. Current intradermal tuberculin skin tests for latent tuberculosis infection (LTBI) based on purified protein derivative (PPD) have poor specificity.Aims. Developing a better skin test antigen as well as a simple skin patch test may improve and facilitate diagnostic performance.Methodology. Defined recombinant antigens that were unique to Mycobacterium tuberculosis (MTB), including two potential latency-associated antigens (ESAT-6 and Rv2653c) and five DosR-encoded latency proteins (Rv1996, Rv2031c, Rv2032, DevR and Rv3716c), were used as diagnostic skin test reagents in comparison with a standard PPD. The performance of the skin tests based on the detection of delayed-type hypersensitivity (DTH) reaction in guinea pigs sensitized to MTB and M. bovis bacille Calmette-Guérin (BCG) vaccine was evaluated.Results. The latency antigens Rv1996, Rv2031c, Rv2032 and Rv2653c and the ESAT-6 protein elicited less reactive DTH skin responses in MTB-sensitized guinea pigs than those resulting from PPD, but elicited no response in BCG-vaccinated guinea pigs. The remaining two latency antigens (DevR and Rv3716c) elicited DTH responses in both groups of animals, as did PPD. The reactivity of PPD in BCG-vaccinated guinea pigs was greater than that of any of the selected skin test reagents. Using stronger concentrations of selected skin test reagents in the patch test led to increased DTH responses that were comparable to those elicited by PPD in guinea pigs sensitized with MTB.Conclusion. Transdermal application of defined purified antigens might be a promising method for LTBI screening.
Collapse
Affiliation(s)
- Songsri Kasempimolporn
- Department of Research and Development, Queen Saovabha Memorial Institute, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Pannatat Areekul
- Department of Research and Development, Queen Saovabha Memorial Institute, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Wichit Thaveekarn
- Department of Research and Development, Queen Saovabha Memorial Institute, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Rattana Sutthisri
- Department of Research and Development, Queen Saovabha Memorial Institute, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Supatsorn Boonchang
- Department of Research and Development, Queen Saovabha Memorial Institute, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Artikaya Sawangvaree
- Department of Research and Development, Queen Saovabha Memorial Institute, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Visith Sitprija
- Department of Research and Development, Queen Saovabha Memorial Institute, Thai Red Cross Society, Bangkok 10330, Thailand
| |
Collapse
|
33
|
Gao X, Wu C, He W, Wang X, Li Y, Wang Y, Jia Y, Yuan R, Li H, Zhang B. DosR antigen Rv1737c induces activation of macrophages dependent on the TLR2 pathway. Cell Immunol 2019; 344:103947. [PMID: 31326120 DOI: 10.1016/j.cellimm.2019.103947] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 06/13/2019] [Accepted: 06/27/2019] [Indexed: 01/22/2023]
Abstract
Latent Mycobacterium tuberculosis (Mtb) infection (LTBI) is the main clinical manifestation after Mtb exposure. During the latent phase, Mtb retards the attempts of eradication by the host immune system. The dormancy survival regulator (DosR) is held as essential for Mtb persistence. Rv1737c is predominantly expressed by the Mtb in latent infection. However, the role of Rv1737c in the immune evasion is still largely unknown. In this study, we have characterized the Rv1737c functions in the recruitment and activation of macrophages, which play a cardinal role in the innate and adaptive immunity. For the first time, we have revealed that Rv1737c induced the tolerogenic phenotype of macrophages by upregulating the expression of indoleamine 2,3-dioxygenase 1 (IDO1). Rv1737c-activated macrophages upregulated interleukin (IL)-4, IL-10, and Foxp3 T cells proliferation in vitro. Furthermore, the interaction of Rv1737c with macrophages was found to depend on the Toll-like receptor 2 (TLR2) pathway. It augmented nuclear factor κB (NF-κB) phosphorylation and co-stimulatory molecule expression. Thus, this study provides a crucial insight into a strategy adopted by Mtb to survive in the host by inducing tolerogenic macrophage expansion.
Collapse
Affiliation(s)
- Xiaoling Gao
- The Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, China.
| | - Cong Wu
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Wenhua He
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Xiaoxia Wang
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Yonghong Li
- The Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Yongxiang Wang
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Yanjuan Jia
- The Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Rui Yuan
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Haojie Li
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Benzhong Zhang
- School of Public Health, Lanzhou University, Lanzhou, China.
| |
Collapse
|
34
|
Singh M, Bhatt P, Sharma M, Varma-Basil M, Chaudhry A, Sharma S. Immunogenicity of late stage specific peptide antigens of Mycobacterium tuberculosis. INFECTION GENETICS AND EVOLUTION 2019; 74:103930. [PMID: 31228643 DOI: 10.1016/j.meegid.2019.103930] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/06/2019] [Accepted: 06/18/2019] [Indexed: 01/05/2023]
Abstract
Global burden of latent TB infection comprises one-third of the world population. Identifying potential Mycobacterium tuberculosis (Mtb) latency associated antigens that can generate protective immunity against the pathogen is crucial for designing an effective TB vaccine. Usually the immune system responds to a small number of amino acids as MHC Class I or Class II peptides. The precision to trigger epitope specific protective T-cell immune response could therefore be achieved with synthetic peptide-based subunit vaccine. In the present study we have considered an immunoinformatic approach using available softwares (ProPred, IEDB, NETMHC, BIMAS, Vaxijen2.0) and docking and visualizing softwares (CABSDOCK, HEX, Pymol, Discovery Studio) to select 10 peptides as latency antigens from 4 proteins (Rv2626, Rv2627, Rv2628, and Rv2032) of DosR regulon of Mtb. As Intracellular IFN-γ secreted by T cells is the most essential cytokine in Th1 mediated protective immunity, these peptides were verified as potential immunogenic epitopes in Peripheral Blood Mononuclear Cells (PBMCs) of 10 healthy contacts of TB patients (HTB) and 10 Category I Pulmonary TB patients (PTB).The antigen-specific CD4 and CD8 T cells expressing intracellular IFN-γ were analyzed using monoclonal antibodies in all subjects by multi-parameter flow cytometry. Both, PTB and HTB individuals responded to DosR peptides by showing increased frequency of IFN-γ+CD4 and IFN-γ+CD8 T cells. The T-cell responses were significantly higher in PTB patients in comparision to the HTB individuals. Additionally, our synthetic peptides and pools showed higher frequencies of IFN-γ+CD4 and IFN-γ+CD8 T cells than the peptides of Ag85B. This pilot study can be taken up further in larger sample size which may support the untapped opportunity of designing Mtb DosR inclusive peptide based post-exposure subunit vaccine.
Collapse
Affiliation(s)
- Medha Singh
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India
| | - Parul Bhatt
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India
| | - Monika Sharma
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India
| | | | - Anil Chaudhry
- Rajan Babu Institute of Pulmonary Medicine and Tuberculosis Hospital, GTB Nagar, Delhi 110009, India
| | - Sadhna Sharma
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| |
Collapse
|
35
|
Halliday A, Masonou T, Tolosa-Wright M, Mandagere V, Lalvani A. Immunodiagnosis of active tuberculosis. Expert Rev Respir Med 2019; 13:521-532. [DOI: 10.1080/17476348.2019.1615888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Alice Halliday
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK
- Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Tereza Masonou
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK
- MRC Centre of Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Mica Tolosa-Wright
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Ajit Lalvani
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
36
|
Liang Y, Zhang X, Bai X, Yang Y, Gong W, Wang T, Ling Y, Zhang J, Wang L, Wang J, Li G, Chen Y, Chen X, Wu X. Immunogenicity and Therapeutic Effects of Latency-Associated Genes in a Mycobacterium Tuberculosis Reactivation Mouse Model. Hum Gene Ther Methods 2019; 30:60-69. [PMID: 30727774 DOI: 10.1089/hgtb.2018.211] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Yan Liang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Xiaoyan Zhang
- Zhengzhou Kingmed Center for Clinical Laboratory, Zhengzhou, P.R. China
| | - Xuejuan Bai
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Yourong Yang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Wenping Gong
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Tong Wang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Yanbo Ling
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Junxian Zhang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Lan Wang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Jie Wang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Gaimei Li
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Yi Chen
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Xiaoyang Chen
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Xueqiong Wu
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis Research, the 8th Medical Center of PLA General Hospital, Beijing, P.R. China
| |
Collapse
|
37
|
Ramaiah A, Nayak S, Rakshit S, Manson AL, Abeel T, Shanmugam S, Sahoo PN, John AJUK, Sundaramurthi JC, Narayanan S, D'Souza G, von Hoegen P, Ottenhoff THM, Swaminathan S, Earl AM, Vyakarnam A. Evidence for Highly Variable, Region-Specific Patterns of T-Cell Epitope Mutations Accumulating in Mycobacterium tuberculosis Strains. Front Immunol 2019; 10:195. [PMID: 30814998 PMCID: PMC6381025 DOI: 10.3389/fimmu.2019.00195] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 01/23/2019] [Indexed: 01/26/2023] Open
Abstract
Vaccines that confer protection through induction of adaptive T-cell immunity rely on understanding T-cell epitope (TCE) evolution induced by immune escape. This is poorly understood in tuberculosis (TB), an ancient, chronic disease, where CD4 T-cell immunity is of recognized importance. We probed 905 functionally validated, curated human CD4 T cell epitopes in 79 Mycobacterium tuberculosis (Mtb) whole genomes from India. This screen resulted in identifying 64 mutated epitopes in these strains initially using a computational pipeline and subsequently verified by single nucleotide polymorphism (SNP) analysis. SNP based phylogeny revealed the 79 Mtb strains to cluster to East African Indian (EAI), Central Asian Strain (CAS), and Beijing (BEI) lineages. Eighty-nine percent of the mutated T-cell epitopes (mTCEs) identified in the 79 Mtb strains from India has not previously been reported. These mTCEs were encoded by genes with high nucleotide diversity scores including seven mTCEs encoded by six antigens in the top 10% of rapidly divergent Mtb genes encoded by these strains. Using a T cell functional assay readout, we demonstrate 62% of mTCEs tested to significantly alter CD4 T-cell IFNγ and/or IL2 secretion with associated changes in predicted HLA-DR binding affinity: the gain of function mutations displayed higher predicted HLA-DR binding affinity and conversely mutations resulting in loss of function displayed lower predicted HLA-DR binding affinity. Most mutated antigens belonged to the cell wall/cell processes, and, intermediary metabolism and respiration families though all known Mtb proteins encoded mutations. Analysis of the mTCEs in an SNP database of 5,310 global Mtb strains identified 82% mTCEs to be significantly more prevalent in Mtb strains isolated from India, including 36 mTCEs identified exclusively in strains from India. These epitopes had a significantly higher predicted binding affinity to HLA-DR alleles that were highly prevalent in India compared to HLA-DR alleles rare in India, highlighting HLA-DR maybe an important driver of these mutations. This first evidence of region-specific TCE mutations potentially employed by Mtb to escape host immunity has important implications for TB vaccine design.
Collapse
Affiliation(s)
- Arunachalam Ramaiah
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Soumya Nayak
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Srabanti Rakshit
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | | | - Thomas Abeel
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | | | - Pravat Nalini Sahoo
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | | | | | - Sujatha Narayanan
- National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | - George D'Souza
- Department of Pulmonary Medicine, St. John's Research Institute, Bangalore, India
| | | | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Ashlee M. Earl
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Annapurna Vyakarnam
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Department of Infectious Diseases, Faculty of Life Sciences & Medicine, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
38
|
Zhao HM, Du R, Li CL, Ji P, Li HC, Wu K, Hu Z, Lu SH, Lowrie DB, Fan XY. Differential T cell responses against DosR-associated antigen Rv2028c in BCG-vaccinated populations with tuberculosis infection. J Infect 2018; 78:275-280. [PMID: 30528871 DOI: 10.1016/j.jinf.2018.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023]
Abstract
The IFN-γ release assays (IGRAs) based on region of difference 1 (RD1) antigens have improved diagnosis of Mycobacterium tuberculosis (Mtb) infection. However, IGRAs with these antigens could not distinguish latent tuberculosis infection (LTBI) from active tuberculosis (ATB). DosR regulon genes are thought to be important for Mtb dormancy, and their products have higher immunogenicity in LTBI than ATB individuals, suggesting protective immunity mediated by DosR regulon-encoded antigens and potential utility of them for differential diagnostics of Mtb-infected populations or development of therapeutic vaccines against tuberculosis (TB). Among them, Rv2028c is a dormancy-related antigen that has demonstrated potential use in TB control, but its immunological characteristics in the BCG-vaccinated Chinese population are unknown. In this study, a total of 148 individuals, including 98 patients with ATB, 20 cases with LTBI and 30 healthy controls, were tested for Rv2028c-specific T cell responses by using an IFN-γ ELISA assay. The results showed that the T-cell responses in LTBI individuals were almost always higher than those in ATB patients, regardless of the site of infection or the results of bacteriological examination in the patients. This allowed for good differentiation between these two groups of Mtb-infected individuals even in the BCG-vaccinated high TB-incidence setting that pertains in China. In addition, the diagnostic efficacy for ATB was enhanced by combining the results from Rv2028c and RD1 antigen-based IFN-γ ELISA assays. In conclusion, Rv2028c-specific T-cell responses might contribute to natural protection against dormant Mtb infection, and the determination of these responses can aid discrimination between healthy LTBI individuals and ATB patients in the Mtb-infected populations.
Collapse
Affiliation(s)
- Hui-Min Zhao
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, 2901 Caolang Rd., Shanghai 201508, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Rui Du
- School of Laboratory Medicine and Life Science, Jinlin Agriculture University, Changchun 130033, China
| | - Chun-Ling Li
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, 2901 Caolang Rd., Shanghai 201508, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Ping Ji
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, 2901 Caolang Rd., Shanghai 201508, China
| | - Hai-Cong Li
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, 2901 Caolang Rd., Shanghai 201508, China
| | - Kang Wu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, 2901 Caolang Rd., Shanghai 201508, China
| | - Zhidong Hu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, 2901 Caolang Rd., Shanghai 201508, China
| | - Shui-Hua Lu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, 2901 Caolang Rd., Shanghai 201508, China
| | - Douglas B Lowrie
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, 2901 Caolang Rd., Shanghai 201508, China
| | - Xiao-Yong Fan
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325035, China; Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, 2901 Caolang Rd., Shanghai 201508, China; School of Laboratory Medicine and Life Science, Jinlin Agriculture University, Changchun 130033, China.
| |
Collapse
|
39
|
Pandey K, Singh S, Bhatt P, Medha, Sharma M, Chaudhry A, Sharma S. DosR proteins of Mycobacterium tuberculosis upregulate effector T cells and down regulate T regulatory cells in TB patients and their healthy contacts. Microb Pathog 2018; 126:399-406. [PMID: 30476579 DOI: 10.1016/j.micpath.2018.11.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 10/27/2022]
Abstract
It is well established that the current problem of tuberculosis (TB) can be combated by overcoming the drawbacks of the currently available BCG vaccine. This would involve incorporation of antigens that can control TB at all stages including the dormant phase which is generally ignored. Hence, DosR regulon proteins, which are expressed in latent infection, could prove to be very good vaccine candidates as they can possibly target the silent but most predominant form of TB infection. In the present study, the immune response to two DosR proteins Rv2627 and Rv2628 has been studied in PBMCs derived from normal individuals, TB patients and healthy contacts of TB patients. It was found that these antigens were capable of stimulating a strong IFN-γ+ T cell response along with accentuation of memory T cells and other protective cytokines such as IL-2 and IL-17. At the same time these proteins decreased the frequencies of immune-suppressor regulatory T cells in in vitro stimulation of PBMC from both patients and their contacts. Considering all these facts together, we suggest Rv2627 and Rv2628 to be one of the extremely promising candidates for incorporation into a post exposure subunit vaccine against TB.
Collapse
Affiliation(s)
- Kirti Pandey
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House, and Department of Zoology, Miranda House, University of Delhi, Delhi, 110007, India.
| | - Swati Singh
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House, and Department of Zoology, Miranda House, University of Delhi, Delhi, 110007, India.
| | - Parul Bhatt
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House, and Department of Zoology, Miranda House, University of Delhi, Delhi, 110007, India.
| | - Medha
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House, and Department of Zoology, Miranda House, University of Delhi, Delhi, 110007, India.
| | - Monika Sharma
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House, and Department of Zoology, Miranda House, University of Delhi, Delhi, 110007, India.
| | - Anil Chaudhry
- Rajan Babu Institute of Pulmonary Medicine and Tuberculosis Hospital, GTB Nagar, Delhi, 110009, India.
| | - Sadhna Sharma
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House, and Department of Zoology, Miranda House, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
40
|
Meier NR, Jacobsen M, Ottenhoff THM, Ritz N. A Systematic Review on Novel Mycobacterium tuberculosis Antigens and Their Discriminatory Potential for the Diagnosis of Latent and Active Tuberculosis. Front Immunol 2018; 9:2476. [PMID: 30473692 PMCID: PMC6237970 DOI: 10.3389/fimmu.2018.02476] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/08/2018] [Indexed: 01/08/2023] Open
Abstract
Background: Current immunodiagnostic tests for tuberculosis (TB) are based on the detection of an immune response toward mycobacterial antigens injected into the skin or following an in-vitro simulation in interferon gamma-release assays. Both tests have limited sensitivity and are unable to differentiate between tuberculosis infection (LTBI) and active tuberculosis disease (aTB). To overcome this, the use of novel Mycobacterium tuberculosis (M. tuberculosis) stage-specific antigens for the diagnosis of LTBI and aTB has gained interest in recent years. This review summarizes current evidence on novel antigens used for the immunodiagnosis of tuberculosis and discrimination of LTBI and aTB. In addition, results on measured biomarkers after stimulation with novel M. tuberculosis antigens were also reviewed. Methods: A systematic literature review was performed in Pubmed, EMBASE and web of science searching articles from 2000 up until December 2017. Only articles reporting studies in humans using novel antigens were included. Results: Of 1,533 articles screened 34 were included in the final analysis. A wide range of novel antigens expressed during different stages and types of LTBI and aTB have been assessed. M. tuberculosis antigens Rv0081, Rv1733c, Rv1737c, Rv2029c, Rv2031 and Rv2628, all encoded by the dormancy of survival regulon, were among the most widely studied antigens and showed the most promising results. These antigens have been shown to have best potential for differentiating LTBI from aTB. In addition, several studies have shown that the inclusion of cytokines other than IFN-γ can improve sensitivity. Conclusion: There is limited evidence that the inclusion of novel antigens as well as the measurement of other biomarkers than IFN-γ may improve sensitivity and may lead to a discrimination of LTBI from aTB.
Collapse
Affiliation(s)
- Noëmi R Meier
- University of Basel Children's Hospital, Mycobacterial Research, Basel, Switzerland.,University of Basel, Faculty of Medicine, Basel, Switzerland
| | - Marc Jacobsen
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Nicole Ritz
- University of Basel Children's Hospital, Mycobacterial Research, Basel, Switzerland.,University of Basel, Faculty of Medicine, Basel, Switzerland.,The Royal Children's Hospital Melbourne, Infectious Disease Unit, Melbourne, VIC, Australia
| |
Collapse
|
41
|
Bai X, Wang D, Liu Y, Xiao L, Liang Y, Yang Y, Zhang J, Lin M, Wu X. Novel epitopes identified from Mycobacterium tuberculosis antigen Rv2629induces cytotoxic T lymphocyte response. Immunol Lett 2018; 203:21-28. [PMID: 29908955 DOI: 10.1016/j.imlet.2018.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 05/11/2018] [Accepted: 06/13/2018] [Indexed: 02/08/2023]
Abstract
There is an urgent need for a more effective vaccine against tuberculosis (TB). Cytotoxic T lymphocytes (CTLs) play a critical role in combating Mycobacterium tuberculosis (M.tb). The identification of novel CTL epitopes is essential for the design of peptide-based vaccines. In this study, we predicted CTL epitope peptides of M.tb antigen Rv2629 restricted by HLA-A2, using bioinformatics methods. The affinity and stability of binding of these peptides with HLA-A2 molecules were detected by flow cytometry. Their ability to induce CTLs generation was determined in peripheral blood mononuclear cells (PBMCs) from healthy uninfected subjects, Latent tuberculosis infection (LTBI) subjects, and TB patients ex vivo. The cytotoxic activity induced by the epitope peptides was tested by lactate dehydrogenase (LDH) release assay. Finally, we found four novel CTL epitope peptides, Rv2629-p190-2L, Rv2629-p190-1Y2L, Rv2629-p274, and Rv2629-p315, which had high-affinity and stability of binding with T2 cells. Their ability of inducing CTLs was highest in PBMCs from TB patients (P < 0.05). In addition, these peptides could induce the CTLs to generate specific cytotoxic activity. They showed higher immunogenicity in TB patients and had the potential to become candidate vaccines for TB therapy.
Collapse
Affiliation(s)
- Xuejuan Bai
- Army Tuberculosis Prevention and Control Key Laboratory, Beijng Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, The 309th Hospital of Chinese PLA, Beijing 100091, PR China
| | - Dongfang Wang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijng Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, The 309th Hospital of Chinese PLA, Beijing 100091, PR China
| | - Yinping Liu
- Army Tuberculosis Prevention and Control Key Laboratory, Beijng Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, The 309th Hospital of Chinese PLA, Beijing 100091, PR China
| | - Li Xiao
- Institute for Organ Transplantation, The 309th Hospital of Chinese PLA, Beijing 100091, PR China
| | - Yan Liang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijng Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, The 309th Hospital of Chinese PLA, Beijing 100091, PR China
| | - Yourong Yang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijng Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, The 309th Hospital of Chinese PLA, Beijing 100091, PR China
| | - Junxian Zhang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijng Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, The 309th Hospital of Chinese PLA, Beijing 100091, PR China
| | - Minggui Lin
- Army Tuberculosis Prevention and Control Key Laboratory, Beijng Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, The 309th Hospital of Chinese PLA, Beijing 100091, PR China
| | - Xueqiong Wu
- Army Tuberculosis Prevention and Control Key Laboratory, Beijng Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, The 309th Hospital of Chinese PLA, Beijing 100091, PR China.
| |
Collapse
|
42
|
Coppola M, Ottenhoff TH. Genome wide approaches discover novel Mycobacterium tuberculosis antigens as correlates of infection, disease, immunity and targets for vaccination. Semin Immunol 2018; 39:88-101. [PMID: 30327124 DOI: 10.1016/j.smim.2018.07.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 07/02/2018] [Indexed: 01/15/2023]
Abstract
Every day approximately six thousand people die of Tuberculosis (TB). Its causative agent, Mycobacterium tuberculosis (Mtb), is an ancient pathogen that through its evolution developed complex mechanisms to evade immune surveillance and acquire the ability to establish persistent infection in its hosts. Currently, it is estimated that one-fourth of the human population is latently infected with Mtb and among those infected 3-10% are at risk of developing active TB disease during their lifetime. The currently available diagnostics are not able to detect this risk group for prophylactic treatment to prevent transmission. Anti-TB drugs are available but only as long regimens with considerable side effects, which could both be reduced if adequate tests were available to monitor the response of TB to treatment. New vaccines are also urgently needed to substitute or boost Bacille Calmette-Guérin (BCG), the only approved TB vaccine: although BCG prevents disseminated TB in infants, it fails to impact the incidence of pulmonary TB in adults, and therefore has little effect on TB transmission. To achieve TB eradication, the discovery of Mtb antigens that effectively correlate with the human response to infection, with the curative host response following TB treatment, and with natural as well as vaccine induced protection will be critical. Over the last decade, many new Mtb antigens have been found and proposed as TB biomarkers and vaccine candidates, but only a very small number of these is being used in commercial diagnostic tests or is being assessed as candidate TB vaccine antigens in human clinical trials, aiming to prevent infection, disease or disease recurrence following treatment. Most of these antigens were discovered decades ago, before the complete Mtb genome sequence became available, and thus did not harness the latest insights from post-genomic antigen discovery strategies and genome wide approaches. These have, for example, revealed critical phase variation in Mtb replication and accompanying gene -and therefore antigen- expression patterns. In this review, we present a brief overview of past methodologies, and subsequently focus on the most important recent Mtb antigen discovery studies which have mined the Mtb antigenome through "unbiased" genome wide approaches. We compare the results for these approaches -as far as we know for the first time-, highlight Mtb antigens that have been identified independently by different strategies and present a comprehensive overview of the Mtb antigens thus discovered.
Collapse
Affiliation(s)
- Mariateresa Coppola
- Dept. Infectious Diseases, LUMC, PO Box 9600, 2300RC Leiden, The Netherlands.
| | - Tom Hm Ottenhoff
- Dept. Infectious Diseases, LUMC, PO Box 9600, 2300RC Leiden, The Netherlands
| |
Collapse
|
43
|
Della Bella C, Spinicci M, Grassi A, Bartalesi F, Benagiano M, Truthmann K, Tapinassi S, Troilo A, D’Elios S, Alnwaisri H, Shuralev E, Singh M, Bartoloni A, D’Elios MM. Novel M. tuberculosis specific IL-2 ELISpot assay discriminates adult patients with active or latent tuberculosis. PLoS One 2018; 13:e0197825. [PMID: 29856871 PMCID: PMC5983515 DOI: 10.1371/journal.pone.0197825] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/09/2018] [Indexed: 12/18/2022] Open
Abstract
Background Tuberculosis (TB) still is a major worldwide health problem, with 10.4 million new cases in 2016. Only 5–15% of people infected with M. tuberculosis develop TB disease while others remain latently infected (LTBI) during their lifetime. Thus, the absence of tests able to distinguish between latent infection and active tuberculosis is one of the major limits of currently available diagnostic tools. Methods A total of 215 patients were included in the study as active TB cases (n = 73), LTBI subjects (n = 88) and healthy persons (n = 54). Peripheral blood mononuclear cells (PBMCs) were isolated from each patient and the LIOSpot® TB anti-human IL-2 ELISpot assay was performed to test their proliferative response to M. tuberculosis antigens ESAT-6, CFP-10 and Ala-DH. Statistical analysis was performed to define the sensitivity and the specificity of the LIOSpot® TB kit for each antigen used and to set the best cut off value that enables discrimination between subjects with active TB or latent TB infection. Results Comparing the LIOSpot® TB results for each tested antigen between uninfected and infected subjects and between people with latent infection and active TB disease, the differences were significant for each antigen (p< 0.0001) but the ROC analysis demonstrated a high accuracy for the Ala-DH test only, with a cut off value of 12.5 SFC per million PBMCs and the Ala-DH ROC curve conferred a 96% sensitivity and 100% specificity to the test. For the ESAT-6 antigen, with a best cut off value of 71.25 SFC per million PBMCs, a sensitivity of 86% and specificity of 36% was obtained. Finally, the best cut off value for CFP-10 was 231.25 SFC per million PBMCs, with a sensitivity of 80% and a specificity of 54%. Thus, as for IGRA assays such as Quantiferon and T-Spot TB tests, ESAT-6 and CFP-10 are unable to distinguish LTBI from active TB when IL-2 is measured. On the contrary, the IL-2 production induced by Ala-DH, measured by LIOSpot® TB kit, shows high sensitivity and specificity for active TB disease. Conclusions This study demonstrates that the LIOSpot® TB test is a highly useful diagnostic tool to discriminate between latent TB infection and active tuberculosis in adults patients.
Collapse
Affiliation(s)
- Chiara Della Bella
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Michele Spinicci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessia Grassi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Filippo Bartalesi
- Infectious and Tropical Diseases Unit, Florence Careggi University Hospital, Florence, Italy
| | - Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Katja Truthmann
- LIONEX Diagnostics and Therapeutics GmbH, Braunschweig, Germany
| | - Simona Tapinassi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Arianna Troilo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Sofia D’Elios
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Heba Alnwaisri
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Eduard Shuralev
- Institute of Environmental Sciences, Kazan Federal University, and Central Research Laboratory, Kazan State Medical Academy, and Federal Center for Toxicological, Radiation and Biological Safety, Kazan, Tatarstan, Russian Federation
| | - Mahavir Singh
- LIONEX Diagnostics and Therapeutics GmbH, Braunschweig, Germany
| | - Alessandro Bartoloni
- Infectious and Tropical Diseases Unit, Florence Careggi University Hospital, Florence, Italy
- * E-mail: (MMDE); (AB)
| | - Mario Milco D’Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- * E-mail: (MMDE); (AB)
| |
Collapse
|
44
|
Meermeier EW, Lewinsohn DM. Early clearance versus control: what is the meaning of a negative tuberculin skin test or interferon-gamma release assay following exposure to Mycobacterium tuberculosis? F1000Res 2018; 7. [PMID: 29904578 PMCID: PMC5974584 DOI: 10.12688/f1000research.13224.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/22/2018] [Indexed: 01/01/2023] Open
Abstract
The elimination of tuberculosis (TB) cannot reasonably be achieved by treatment of individual cases and will require an improved vaccine or immunotherapy. A challenge in developing an improved TB vaccine has been the lack of understanding what is needed to generate sterilizing immunity against
Mycobacterium tuberculosis (Mtb) infection. Several epidemiological observations support the hypothesis that humans can eradicate Mtb following exposure. This has been termed early clearance and is defined as elimination of Mtb infection prior to the development of an adaptive immune response, as measured by a tuberculin skin test or interferon-gamma release assay. Here, we examine research into the likelihood of and possible mechanisms responsible for early clearance in household contacts of patients with active TB. We explore both innate and adaptive immune responses in the lung. Enhanced understanding of these mechanisms could be harnessed for the development of a preventative vaccine or immunotherapy.
Collapse
Affiliation(s)
- Erin W Meermeier
- Pulmonary and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, USA
| | - David M Lewinsohn
- Pulmonary and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, USA.,Department of Medicine, VA Portland Health Care System, Portland, OR, USA
| |
Collapse
|
45
|
Orlando V, La Manna MP, Goletti D, Palmieri F, Lo Presti E, Joosten SA, La Mendola C, Buccheri S, Ottenhoff THM, Dieli F, Caccamo N. Human CD4 T-Cells With a Naive Phenotype Produce Multiple Cytokines During Mycobacterium Tuberculosis Infection and Correlate With Active Disease. Front Immunol 2018; 9:1119. [PMID: 29875774 PMCID: PMC5974168 DOI: 10.3389/fimmu.2018.01119] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/03/2018] [Indexed: 12/21/2022] Open
Abstract
T-cell-mediated immune responses play a fundamental role in controlling Mycobacterium tuberculosis (M. tuberculosis) infection, and traditionally, this response is thought to be mediated by Th1-type CD4+ T-cells secreting IFN-γ. While studying the function and specificity of M. tuberculosis-reactive CD4+ T-cells in more detail at the single cell level; however, we found a human CD4+ T-cell population with a naive phenotype that interestingly was capable of producing multiple cytokines (TCNP cells). CD4+ TCNP cells phenotyped as CD95lo CD28int CD49dhi CXCR3hi and showed a broad distribution of T cell receptor Vβ segments. They rapidly secreted multiple cytokines in response to different M. tuberculosis antigens, their frequency was increased during active disease, but was comparable to latent tuberculosis infection in treated TB patients. These results identify a novel human CD4+ T-cell subset involved in the human immune response to mycobacteria, which is present in active TB patients’ blood. These results significantly expand our understanding of the immune response in infectious diseases.
Collapse
Affiliation(s)
- Valentina Orlando
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Biopathology, University of Palermo, Palermo, Italy
| | - Marco P La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Biopathology, University of Palermo, Palermo, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases L. Spallanzani, Rome, Italy
| | - Fabrizio Palmieri
- Translational Research Unit, National Institute for Infectious Diseases L. Spallanzani, Rome, Italy
| | - Elena Lo Presti
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Biopathology, University of Palermo, Palermo, Italy
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Simona Buccheri
- Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione, Palermo, Italy
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Biopathology, University of Palermo, Palermo, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Biopathology, University of Palermo, Palermo, Italy
| |
Collapse
|
46
|
Zhu B, Dockrell HM, Ottenhoff THM, Evans TG, Zhang Y. Tuberculosis vaccines: Opportunities and challenges. Respirology 2018; 23:359-368. [PMID: 29341430 DOI: 10.1111/resp.13245] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/12/2017] [Accepted: 12/04/2017] [Indexed: 12/11/2022]
Abstract
Tuberculosis (TB) is a serious disease around the world. Bacillus Calmette-Guérin (BCG) is the only TB vaccine licensed for use in human beings, and is effective in protecting infants and children against severe miliary and meningeal TB. However, BCG's protective efficacy is variable in adults. Novel TB vaccine candidates being developed include whole-cell vaccines (recombinant BCG (rBCG), attenuated Mycobacterium tuberculosis, killed M. tuberculosis or Mycobacterium vaccae), adjuvanted protein subunit vaccines, viral vector-delivered subunit vaccines, plasmid DNA vaccines, RNA-based vaccines etc. At least 12 novel TB vaccine candidates are now in clinical trials, including killed M. vaccae, rBCG ΔureC::hly, adjuvanted fusion proteins M72 and H56 and viral vectored MVA85A. Unfortunately, in TB, there are no correlates of vaccine-induced protection, although cell-mediated immune responses such as interferon-gamma (IFN-γ) production are widely used to assess vaccine's immunogenicity. Recent studies suggested that central memory T cells and local secreted IgA correlated with protection against TB disease. Clinical TB vaccine efficacy trials should invest in identifying correlates of protection, and evaluate new TB biomarkers emerging from human and animal studies. Accumulating new knowledge on M. tuberculosis antigens and immune profiles correlating with protection or disease risk will be of great help in designing next generation of TB vaccines.
Collapse
Affiliation(s)
- Bingdong Zhu
- Lanzhou Center for Tuberculosis Research and Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Hazel M Dockrell
- Department of Immunology and Infection and Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, UK
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
47
|
Arroyo L, Marín D, Franken KLMC, Ottenhoff THM, Barrera LF. Potential of DosR and Rpf antigens from Mycobacterium tuberculosis to discriminate between latent and active tuberculosis in a tuberculosis endemic population of Medellin Colombia. BMC Infect Dis 2018; 18:26. [PMID: 29310595 PMCID: PMC5759254 DOI: 10.1186/s12879-017-2929-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 12/17/2017] [Indexed: 01/07/2023] Open
Abstract
Background Tuberculosis (TB) remains one of the most deadly infectious diseases. One-third to one-fourth of the human population is estimated to be infected with Mycobacterium tuberculosis (Mtb) without showing clinical symptoms, a condition called latent TB infection (LTBI). Diagnosis of Mtb infection is based on the immune response to a mixture of mycobacterial antigens (PPD) or to Mtb specific ESAT-6/CFP10 antigens (IGRA), highly expressed during the initial phase of infection. However, the immune response to PPD and IGRA antigens has a low power to discriminate between LTBI and PTB. The T-cell response to a group of so-called latency (DosR-regulon-encoded) and Resuscitation Promoting (Rpf) antigens of Mtb has been proved to be significantly higher in LTBI compared to active TB across many populations, suggesting their potential use as biomarkers to differentiate latent from active TB. Methods PBMCs from a group LTBI (n = 20) and pulmonary TB patients (PTB, n = 21) from an endemic community for TB of the city of Medellín, Colombia, were in vitro stimulated for 7 days with DosR- (Rv1737c, Rv2029c, and Rv2628), Rpf- (Rv0867c and Rv2389c), the recombinant fusion protein ESAT-6-CFP10 (E6-C10)-, or PPD-antigen. The induced IFNγ levels detectable in the supernatants of the antigen-stimulated cells were then used to calculate specificity and sensitivity in discriminating LTBI from PTB, using different statistical approaches. Results IFNγ production in response to DosR and Rpf antigens was significantly higher in LTBI compared to PTB. ROC curve analyses of IFNγ production allowed differentiation of LTBI from PTB with areas under the curve higher than 0.70. Furthermore, Multiple Correspondence Analysis (MCA) revealed that LTBI is associated with higher levels of IFNγ in response to the different antigens compared to PTB. Analysis based on decision trees showed that the IFNγ levels produced in response to Rv2029c was the leading variable that best-classified disease status. Finally, logistic regression analysis predicted that IFNγ produced by PBMCs in response to E6-C10, Rv2029c, Rv0867c (RpfA) and Rv2389c (RpfA) antigens correlates best with the probability of being latently infected. Conclusions The Mtb antigens E6-C10, Rv2029c (PfkB), Rv0867c (RpfA) and Rv2389c (RpfA), may be potential candidates to discriminate LTBI from PTB. Electronic supplementary material The online version of this article (doi: 10.1186/s12879-017-2929-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leonar Arroyo
- Grupo de Inmunología Celular e Inmunogenética (GICIG), Albinusdreef 2, 2333, Leiden, ZA, Netherlands
| | - Diana Marín
- Universidad Pontificia Bolivariana (UPB), Albinusdreef 2, 2333, Leiden, ZA, Netherlands
| | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Centre, Albinusdreef 2, 2333, Leiden, ZA, Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Centre, Albinusdreef 2, 2333, Leiden, ZA, Netherlands
| | - Luis F Barrera
- Grupo de Inmunología Celular e Inmunogenética (GICIG), Albinusdreef 2, 2333, Leiden, ZA, Netherlands. .,Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia, UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
48
|
de Araujo LS, da Silva NDBM, Leung JAM, Mello FCQ, Saad MHF. IgG subclasses' response to a set of mycobacterial antigens in different stages of Mycobacterium tuberculosis infection. Tuberculosis (Edinb) 2018. [DOI: 10.1016/j.tube.2017.10.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
49
|
Pathakumari B, Devasundaram S, Raja A. Altered expression of antigen-specific memory and regulatory T-cell subsets differentiate latent and active tuberculosis. Immunology 2017; 153:325-336. [PMID: 28881482 DOI: 10.1111/imm.12833] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 08/15/2017] [Accepted: 08/31/2017] [Indexed: 01/20/2023] Open
Abstract
Although one-third of the world population is infected with Mycobacterium tuberculosis, only 5-10% of the infected individuals will develop active tuberculosis (TB) disease and the rest will remain infected with no symptoms, known as latent TB infection (LTBI). Identifying biomarkers that differentiate latent and active TB disease enables effective TB control, as early detection, treatment of active TB and preventive treatment of individuals with LTBI are crucial steps involved in TB control. Here, we have evaluated the frequency of antigen-specific memory and regulatory T (Treg) cells in 15 healthy household contacts (HHC) and 15 pulmonary TB patients (PTB) to identify biomarkers for differential diagnosis of LTBI and active TB. Among all the antigens tested in the present study, early secretory antigenic target-6 (ESAT-6) -specific CD4+ and CD8+ central memory (Tcm) cells showed 93% positivity in HHC and 20% positivity in PTB. The novel test antigens Rv0753c and Rv0009 both displayed 80% and 20% positivity in HHC and PTB, respectively. In contrast to Tcm cells, effector memory T (Tem) cells showed a higher response in PTB than HHC; both ESAT-6 and Rv0009 showed similar positivity of 80% in PTB and 33% in HHC. PTB patients have a higher proportion of circulating antigen-reactive Treg cells (CD4+ CD25+ FoxP3+ ) than LTBI. Rv2204c-specific Treg cells showed maximum positivity of 73% in PTB and 20% in HHC. Collectively, our data conclude that ESAT-6-specific Tcm cells and Rv2204c-specific Treg cells might be useful biomarkers to discriminate LTBI from active TB.
Collapse
Affiliation(s)
- Balaji Pathakumari
- Department of Immunology, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | - Santhi Devasundaram
- Department of Immunology, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | - Alamelu Raja
- Department of Immunology, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| |
Collapse
|
50
|
Anaerobic Mycobacterium tuberculosis Cell Death Stems from Intracellular Acidification Mitigated by the DosR Regulon. J Bacteriol 2017; 199:JB.00320-17. [PMID: 28874407 DOI: 10.1128/jb.00320-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/23/2017] [Indexed: 01/28/2023] Open
Abstract
Mycobacterium tuberculosis is a strict aerobe capable of prolonged survival in the absence of oxygen. We investigated the ability of anaerobic M. tuberculosis to counter challenges to internal pH homeostasis in the absence of aerobic respiration, the primary mechanism of proton efflux for aerobic bacilli. Anaerobic M. tuberculosis populations were markedly impaired for survival under a mildly acidic pH relative to standard culture conditions. An acidic environmental pH greatly increased the susceptibilities of anaerobic bacilli to the collapse of the proton motive force by protonophores, to antimicrobial compounds that target entry into the electron transport system, and to small organic acids with uncoupling activity. However, anaerobic bacilli exhibited high tolerance against these challenges at a near-neutral pH. At a slightly alkaline pH, which was near the optimum intracellular pH, the addition of protonophores even improved the long-term survival of bacilli. Although anaerobic M. tuberculosis bacilli under acidic conditions maintained 40% lower ATP levels than those of bacilli under standard culture conditions, ATP loss alone could not explain the drop in viability. Protonophores decreased ATP levels by more than 90% regardless of the extracellular pH but were bactericidal only under acidic conditions, indicating that anaerobic bacilli could survive an extreme ATP loss provided that the external pH was within viable intracellular parameters. Acidic conditions drastically decreased the anaerobic survival of a DosR mutant, while an alkaline environment improved the survival of the DosR mutant. Together, these findings indicate that intracellular acidification is a primary challenge for the survival of anaerobic M. tuberculosis and that the DosR regulon plays a critical role in sustaining internal pH homeostasis.IMPORTANCE During infection, M. tuberculosis bacilli are prevalent in environments largely devoid of oxygen, yet the factors that influence the survival of these severely growth-limited and metabolically limited bacilli remain poorly understood. We determined how anaerobic bacilli respond to fluctuations in environmental pH and observed that these bacilli were highly susceptible to stresses that promoted internal acidic stress, whereas conditions that promoted an alkaline internal pH promoted long-term survival even during severe ATP depletion. The DosR regulon, a major regulator of general hypoxic stress, played an important role in maintaining internal pH homeostasis under anaerobic conditions. Together, these findings indicate that in the absence of aerobic respiration, protection from internal acidification is crucial for long-term M. tuberculosis survival.
Collapse
|