101
|
Ahmad S, Bhattacharya D, Gupta N, Rawat V, Tousif S, Van Kaer L, Das G. Clofazimine enhances the efficacy of BCG revaccination via stem cell-like memory T cells. PLoS Pathog 2020; 16:e1008356. [PMID: 32437421 PMCID: PMC7269335 DOI: 10.1371/journal.ppat.1008356] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/03/2020] [Accepted: 01/27/2020] [Indexed: 12/11/2022] Open
Abstract
Tuberculosis (TB) is one of the deadliest diseases, claiming ~2 million deaths annually worldwide. The majority of people in TB endemic regions are vaccinated with Bacillus Calmette Guerin (BCG), which is the only usable vaccine available. BCG is efficacious against meningeal and disseminated TB in children, but protective responses are relatively short-lived and fail to protect against adult pulmonary TB. The longevity of vaccine efficacy critically depends on the magnitude of long-lasting central memory T (TCM) cells, a major source of which is stem cell-like memory T (TSM) cells. These TSM cells exhibit enhanced self-renewal capacity as well as to rapidly respond to antigen and generate protective poly-functional T cells producing IFN-γ, TNF-α, IL-2 and IL-17. It is now evident that T helper Th 1 and Th17 cells are essential for host protection against TB. Recent reports have indicated that Th17 cells preserve the molecular signature for TSM cells, which eventually differentiate into IFN-γ-producing effector cells. BCG is ineffective in inducing Th17 cell responses, which might explain its inadequate vaccine efficacy. Here, we show that revaccination with BCG along with clofazimine treatment promotes TSM differentiation, which continuously restores TCM and T effector memory (TEM) cells and drastically increases vaccine efficacy in BCG-primed animals. Analyses of these TSM cells revealed that they are predominantly precursors to host protective Th1 and Th17 cells. Taken together, these findings revealed that clofazimine treatment at the time of BCG revaccination provides superior host protection against TB by increasing long-lasting TSM cells. Tuberculosis (TB) is one of the deadliest diseases, claiming ~2 million deaths annually worldwide. Bacillus Calmette Guerin (BCG) is the only usable vaccine available and exhibits efficacy against meningeal and disseminated TB in children. Consequently, the vast majority of people in TB endemic regions are vaccinated with BCG. However, host protective immune responses diminish over time due to gradual depletion of T central memory (TCM) cells, which are responsible for long-term host protection. Here, we provide evidence that revaccination with BCG along with the clofazimine, an approved drug for treatment of leprosy and drug-resistant TB, induces stem cell-like memory T (TSM) cells. TSM cells are precursors to TCM cells, and provide long-term host protection to TB by continuous supply of TCM cells. Interestingly, these TSM cells were generated from IL-17-producing T helper (Th)17 cells. These TSM cells differentiated into TCM and T effector memory (TEM) cells and maintained a stable pool of critically important Th1 and Th17 cells to provide optimal host protection against TB.
Collapse
Affiliation(s)
- Shaheer Ahmad
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | | | - Neeta Gupta
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Varsha Rawat
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Sultan Tousif
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Gobardhan Das
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
- * E-mail:
| |
Collapse
|
102
|
Künzli M, King CG. Resident Memory T Cells Escape 'Home Quarantine'. Trends Immunol 2020; 41:454-456. [PMID: 32386961 DOI: 10.1016/j.it.2020.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/16/2022]
Abstract
T resident memory (Trm) cells provide a first line of defense in non-lymphoid tissues. A new report in Nature Immunology by Fonseca et al. reveals that CD8+ Trm cells can give rise to circulating effector and memory T cells, but remain predisposed to migrate back into their tissue of origin.
Collapse
Affiliation(s)
- Marco Künzli
- Department of Biomedicine, University of Basel, University Hospital of Basel, CH-4031 Basel, Switzerland
| | - Carolyn G King
- Department of Biomedicine, University of Basel, University Hospital of Basel, CH-4031 Basel, Switzerland.
| |
Collapse
|
103
|
Chai Q, Lu Z, Liu CH. Host defense mechanisms against Mycobacterium tuberculosis. Cell Mol Life Sci 2020; 77:1859-1878. [PMID: 31720742 PMCID: PMC11104961 DOI: 10.1007/s00018-019-03353-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/30/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022]
Abstract
Tuberculosis (TB), which is caused by Mycobacterium tuberculosis (Mtb), remains the leading cause of death worldwide from a single infectious pathogen. Mtb is a paradigmatic intracellular pathogen that primarily invades the lungs after host inhalation of bacteria-containing droplets via the airway. However, the majority of Mtb-exposed individuals can spontaneously control the infection by virtue of a robust immune defense system. The mucosal barriers of the respiratory tract shape the first-line defense against Mtb through various mucosal immune responses. After arriving at the alveoli, the surviving mycobacteria further encounter a set of host innate immune cells that exert multiple cellular bactericidal functions. Adaptive immunity, predominantly mediated by a range of different T cell and B cell subsets, is subsequently activated and participates in host anti-mycobacterial defense. During Mtb infection, host bactericidal immune responses are exquisitely adjusted and balanced by multifaceted mechanisms, including genetic and epigenetic regulation, metabolic regulation and neuroendocrine regulation, which are indispensable for maintaining host immune efficiency and avoiding excessive tissue injury. A better understanding of the integrated and equilibrated host immune defense system against Mtb will contribute to the development of rational TB treatment regimens especially novel host-directed therapeutics.
Collapse
Affiliation(s)
- Qiyao Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Zhe Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
104
|
Tim-3: A co-receptor with diverse roles in T cell exhaustion and tolerance. Semin Immunol 2020; 42:101302. [PMID: 31604535 DOI: 10.1016/j.smim.2019.101302] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 08/30/2019] [Indexed: 12/13/2022]
Abstract
T cell inhibitory co-receptors play a crucial role in maintaining the balance between physiologic immune responses and maladaptive ones. T cell immunoglobulin and mucin domain-containing-3 (Tim-3) is a unique inhibitory co-receptor in that its expression is chiefly restricted to interferon (IFN)γ-producing CD4+ and CD8+ T cells. Early reports firmly established its importance in maintaining peripheral tolerance in transplantation and autoimmunity. However, it has become increasingly clear that Tim-3 expression on T cells, together with other check-point molecules, in chronic infections and cancers can hinder productive immune responses. In this review, we outline what is currently known about the regulation of Tim-3 expression, its ligands and signaling. We discuss both its salutary and deleterious function in immune disorders, as well as the T cell-extrinsic and -intrinsic factors that regulate its function.
Collapse
|
105
|
Abstract
Tuberculosis (TB) vaccine research has reached a unique point in time. Breakthrough findings in both the basic immunology of Mycobacterium tuberculosis infection and the clinical development of TB vaccines suggest, for the first time since the discovery of the Mycobacterium bovis bacillus Calmette-Guérin (BCG) vaccine more than a century ago, that a novel, efficacious TB vaccine is imminent. Here, we review recent data in the light of our current understanding of the immunology of TB infection and discuss the identification of biomarkers for vaccine efficacy and the next steps in the quest for an efficacious vaccine that can control the global TB epidemic.
Collapse
|
106
|
Young C, Walzl G, Du Plessis N. Therapeutic host-directed strategies to improve outcome in tuberculosis. Mucosal Immunol 2020; 13:190-204. [PMID: 31772320 PMCID: PMC7039813 DOI: 10.1038/s41385-019-0226-5] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/15/2019] [Accepted: 10/23/2019] [Indexed: 02/04/2023]
Abstract
Bacille Calmette-Guérin (BCG) is the only licenced tuberculosis (TB) vaccine, but has limited efficacy against pulmonary TB disease development and modest protection against extrapulmonary TB. Preventative antibiotic treatment for Mycobacterium tuberculosis (Mtb) infections in high-prevalence settings is unfeasible due to unclear treatment durability, drug toxicity, logistical constraints related to directly observed treatment strategy (DOTS) and the lengthy treatment protocols. Together, these factors promote non-adherence, contributing to relapse and establishment of drug-resistant Mtb strains. Although antibiotic treatment of drug-susceptible Mtb is generally effective, drug-resistant TB has a treatment efficacy below 50% and can, in a proportion, develop into progressive, untreatable disease. Other immune compromising co-infections and/or co-morbidities require more complex prevention/treatment approaches, posing huge financial burdens to national health services. Novel TB treatment strategies, such as host-directed therapeutics, are required to complement pathogen-targeted approaches. Pre-clinical studies have highlighted promising candidates that enhance endogenous pathways and/or limit destructive host responses. This review discusses promising pre-clinical candidates and forerunning compounds at advanced stages of clinical investigation in TB host-directed therapeutic (HDT) efficacy trials. Such approaches are rationalized to improve outcome in TB and shorten treatment strategies.
Collapse
Affiliation(s)
- C Young
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology/DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - G Walzl
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology/DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - N Du Plessis
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology/DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
107
|
Abstract
Tuberculosis (TB) is a serious global public health challenge that results in significant morbidity and mortality worldwide. TB is caused by infection with the bacilli Mycobacterium tuberculosis (M. tuberculosis), which has evolved a wide variety of strategies in order to thrive within its host. Understanding the complex interactions between M. tuberculosis and host immunity can inform the rational design of better TB vaccines and therapeutics. This chapter covers innate and adaptive immunity against M. tuberculosis infection, including insights on bacterial immune evasion and subversion garnered from animal models of infection and human studies. In addition, this chapter discusses the immunology of the TB granuloma, TB diagnostics, and TB comorbidities. Finally, this chapter provides a broad overview of the current TB vaccine pipeline.
Collapse
|
108
|
Barber DL, Sakai S, Kudchadkar RR, Fling SP, Day TA, Vergara JA, Ashkin D, Cheng JH, Lundgren LM, Raabe VN, Kraft CS, Nieva JJ, Cheever MA, Nghiem PT, Sharon E. Tuberculosis following PD-1 blockade for cancer immunotherapy. Sci Transl Med 2020; 11:11/475/eaat2702. [PMID: 30651320 DOI: 10.1126/scitranslmed.aat2702] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 10/02/2018] [Accepted: 12/06/2018] [Indexed: 12/11/2022]
Abstract
Because of the well-established therapeutic benefit of boosting antitumor responses through blockade of the T cell inhibitory receptor PD-1, it has been proposed that PD-1 blockade could also be useful in infectious disease settings, including Mycobacterium tuberculosis (Mtb) infection. However, in preclinical models, Mtb-infected PD-1-/- mice mount exaggerated TH1 responses that drive lethal immunopathology. Multiple cases of tuberculosis during PD-1 blockade have been observed in patients with cancer, but in humans little is understood about Mtb-specific immune responses during checkpoint blockade-associated tuberculosis. Here, we report two more cases. We describe a patient who succumbed to disseminated tuberculosis after PD-1 blockade for treatment of nasopharyngeal carcinoma, and we examine Mtb-specific immune responses in a patient with Merkel cell carcinoma who developed checkpoint blockade-associated tuberculosis and was successfully treated for the infection. After anti-PD-1 administration, interferon-γ-producing Mtb-specific CD4 T cells became more prevalent in the blood, and a tuberculoma developed a few months thereafter. Mtb-specific TH17 cells, CD8 T cells, regulatory T cells, and antibody abundance did not change before the appearance of the granuloma. These results are consistent with the murine model data and suggest that boosting TH1 function with PD-1 blockade may increase the risk or severity of tuberculosis in humans.
Collapse
Affiliation(s)
- Daniel L Barber
- T Lymphocyte Biology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA.
| | - Shunsuke Sakai
- T Lymphocyte Biology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Ragini R Kudchadkar
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Steven P Fling
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Tracey A Day
- Clinical Immunology Group, Infectious Disease Research Institute, Seattle, WA 98102, USA
| | - Julie A Vergara
- Clinical Immunology Group, Infectious Disease Research Institute, Seattle, WA 98102, USA
| | - David Ashkin
- Division of Infectious Diseases and Global Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Jonathan H Cheng
- Norris Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Lisa M Lundgren
- Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Vanessa N Raabe
- Division of Infectious Diseases, Emory University, Atlanta, GA 30322, USA
| | - Colleen S Kraft
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, GA 30322, USA
| | - Jorge J Nieva
- Norris Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Martin A Cheever
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Paul T Nghiem
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Elad Sharon
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
109
|
Hepatitis C Virus Affects Tuberculosis-Specific T Cells in HIV-Negative Patients. Viruses 2020; 12:v12010101. [PMID: 31952232 PMCID: PMC7019953 DOI: 10.3390/v12010101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 12/28/2019] [Accepted: 01/08/2020] [Indexed: 12/13/2022] Open
Abstract
The occurrence of tuberculosis (TB) and hepatitis C virus (HCV) infections in the same patient presents a unique clinical challenge. The impact of HCV infection on the immune response to TB remains poorly investigated in TB+/HCV+ patients. This study was conducted to evaluate the impact of HCV on the T-cell-mediated immune response to TB in coinfected patients. Sixty-four patients with active TB infections were screened for coinfection with HCV. The expression of immune activation markers IFN-γ, CD38, and HLA-DR on TB-specific CD4+ T cells was evaluated by flow cytometry in TB-monoinfected patients, TB/HCV-coinfected patients, and healthy controls. IL-2, IL-4, IFN-γ, TNF-α, and IL-10 levels were measured using ELISA. The end-of-treatment response to anti-TB therapy was recorded for both patient groups. Significantly lower levels of CD4+IFN-γ+CD38+ and CD4+IFN-γ+HLA-DR+ T cells were detected in TB/HCV-coinfected patients compared to TB monoinfected patients and controls. TB+/HCV+-coinfected patients showed higher serum levels of IL-10. The baseline frequencies of TB-specific activated T-cell subsets did not predict the response to antituberculous therapy in TB+/HCV+ patients. We concluded that different subsets of TB-specific CD4+ T cells in TB/HCV-infected individuals are partially impaired in early-stage HCV infection. This was combined with increased serum IL-10 level. Such immune modulations may represent a powerful risk factor for disease progression in patients with HCV/TB coinfection.
Collapse
|
110
|
Hope JL, Spantidea PI, Kiernan CH, Stairiker CJ, Rijsbergen LC, van Meurs M, Brouwers-Haspels I, Mueller YM, Nelson DJ, Bradley LM, Aerts JGJV, Katsikis PD. Microenvironment-Dependent Gradient of CTL Exhaustion in the AE17sOVA Murine Mesothelioma Tumor Model. Front Immunol 2020; 10:3074. [PMID: 31998326 PMCID: PMC6968785 DOI: 10.3389/fimmu.2019.03074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 12/16/2019] [Indexed: 01/26/2023] Open
Abstract
The immune system, and in particular, cytotoxic CD8+ T cells (CTLs), plays a vital part in the prevention and elimination of tumors. In many patients, however, CTL-mediated tumor killing ultimately fails in the clearance of cancer cells resulting in disease progression, in large part due to the progression of effector CTL into exhausted CTL. While there have been major breakthroughs in the development of CTL-mediated “reinvigoration”-driven immunotherapies such as checkpoint blockade therapy, there remains a need to better understand the drivers behind the development of T cell exhaustion. Our study highlights the unique differences in T cell exhaustion development in tumor-specific CTL which arises over time in a mouse model of mesothelioma. Importantly, we also show that peripheral tumor-specific T cells have a unique expression profile compared to exhausted tumor-infiltrating CTL at a late-stage of tumor progression in mice. Together, these data suggest that greater emphasis should be placed on understanding contributions of individual microenvironments in the development of T cell exhaustion.
Collapse
Affiliation(s)
- Jennifer L Hope
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Cancer Immunology and Tumor Microenvironment Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Panagiota I Spantidea
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Caoimhe H Kiernan
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | | | - Laurine C Rijsbergen
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Marjan van Meurs
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Inge Brouwers-Haspels
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Yvonne M Mueller
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Delia J Nelson
- Immunology and Cancer Group, School of Biomedical Sciences, Curtin University, Perth, WA, Australia
| | - Linda M Bradley
- Cancer Immunology and Tumor Microenvironment Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Peter D Katsikis
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
111
|
Patankar YR, Sutiwisesak R, Boyce S, Lai R, Lindestam Arlehamn CS, Sette A, Behar SM. Limited recognition of Mycobacterium tuberculosis-infected macrophages by polyclonal CD4 and CD8 T cells from the lungs of infected mice. Mucosal Immunol 2020; 13:140-148. [PMID: 31636345 PMCID: PMC7161428 DOI: 10.1038/s41385-019-0217-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/23/2019] [Accepted: 09/28/2019] [Indexed: 02/04/2023]
Abstract
Immune responses following Mycobacterium tuberculosis (Mtb) infection or vaccination are frequently assessed by measuring T-cell recognition of crude Mtb antigens, recombinant proteins, or peptide epitopes. We previously showed that not all Mtb-specific T cells recognize Mtb-infected macrophages. Thus, an important question is what proportion of T cells elicited by Mtb infection recognize Mtb-infected macrophages. We address this question by developing a modified elispot assay using viable Mtb-infected macrophages, a low multiplicity of infection and purified T cells. In C57BL/6 mice, CD4 and CD8 T cells were classically MHC restricted. Comparable frequencies of T cells that recognize Mtb-infected macrophages were determined using interferon-γ elispot and intracellular cytokine staining, and lung CD4 T cells more sensitively recognized Mtb-infected macrophages than lung CD8 T cells. Compared to the relatively high frequencies of T cells specific for antigens such as ESAT-6 and TB10.4, low frequencies of total pulmonary T cells elicited by aerosolized Mtb infection recognize Mtb-infected macrophages. Finally, we demonstrate that BCG vaccination elicits T cells that recognize Mtb-infected macrophages. We propose that the frequency of T cells that recognize infected macrophages could correlate with protective immunity and may be an alternative approach to measuring T-cell responses to Mtb antigens.
Collapse
Affiliation(s)
- Yash R. Patankar
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Rujapak Sutiwisesak
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Shayla Boyce
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Rocky Lai
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Cecilia S. Lindestam Arlehamn
- 0000 0004 0461 3162grid.185006.aDepartment of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Alessandro Sette
- 0000 0004 0461 3162grid.185006.aDepartment of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037 USA ,0000 0001 2107 4242grid.266100.3Department of Medicine, University of California San Diego, La Jolla, CA 92093 USA
| | - Samuel M. Behar
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| |
Collapse
|
112
|
Hakim MS, Rahmadika N, Jariah ROA. Expressions of inhibitory checkpoint molecules in acute and chronic HBV and HCV infections: Implications for therapeutic monitoring and personalized therapy. Rev Med Virol 2019; 30:e2094. [DOI: 10.1002/rmv.2094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Mohamad S. Hakim
- Department of Microbiology, Faculty of Medicine, Public Health and NursingUniversitas Gadjah Mada Yogyakarta Indonesia
| | - Nofri Rahmadika
- Infectious Disease Research Center, Faculty of MedicineUniversitas Padjadjaran Bandung Indonesia
| | - Rizka O. A. Jariah
- Department of Health Science, Faculty of Vocational StudiesUniversitas Airlangga Surabaya Indonesia
| |
Collapse
|
113
|
Abstract
Tuberculosis (TB) is the leading killer among all infectious diseases worldwide despite extensive use of the Mycobacterium bovis bacille Calmette-Guérin (BCG) vaccine. A safer and more effective vaccine than BCG is urgently required. More than a dozen TB vaccine candidates are under active evaluation in clinical trials aimed to prevent infection, disease, and recurrence. After decades of extensive research, renewed promise of an effective vaccine against this ancient airborne disease has recently emerged. In two innovative phase 2b vaccine clinical trials, one for the prevention of Mycobacterium tuberculosis infection in healthy adolescents and another for the prevention of TB disease in M. tuberculosis-infected adults, efficacy signals were observed. These breakthroughs, based on the greatly expanded knowledge of the M. tuberculosis infection spectrum, immunology of TB, and vaccine platforms, have reinvigorated the TB vaccine field. Here, we review our current understanding of natural immunity to TB, limitations in BCG immunity that are guiding vaccinologists to design novel TB vaccine candidates and concepts, and the desired attributes of a modern TB vaccine. We provide an overview of the progress of TB vaccine candidates in clinical evaluation, perspectives on the challenges faced by current vaccine concepts, and potential avenues to build on recent successes and accelerate the TB vaccine research-and-development trajectory.
Collapse
|
114
|
Liu X, Li F, Niu H, Ma L, Chen J, Zhang Y, Peng L, Gan C, Ma X, Zhu B. IL-2 Restores T-Cell Dysfunction Induced by Persistent Mycobacterium tuberculosis Antigen Stimulation. Front Immunol 2019; 10:2350. [PMID: 31632413 PMCID: PMC6783502 DOI: 10.3389/fimmu.2019.02350] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 09/17/2019] [Indexed: 12/18/2022] Open
Abstract
Tuberculosis (TB) is a chronic disease mainly caused by Mycobacterium tuberculosis. The function of T cells usually decreased and even exhausted in severe TB such as multiple drug resistant TB (MDR-TB), which might lead to the failure of treatment in return. The mechanism of T cell dysfunction in TB is still not clear. In this study we set up a mouse model of T cell dysfunction by persistent M. tuberculosis antigen stimulation and investigated the therapeutic role of interleukin 2 (IL-2) in it. C57BL/6 mice were primed with Mycobacterium bovis Bacillus Calmette-Guérin (BCG) and boosted repeatedly with a combination of M. tuberculosis fusion proteins Mtb10.4-HspX (MH) plus ESAT6-Ag85B-MPT64 <190-198>-Mtb8.4-Rv2626c (LT70) or MH plus ESAT6 and CFP10 with adjuvant of N, N'-dimethyl-N, N'-dioctadecylammonium bromide (DDA) plus polyinosinic-polycytidylic acid (Poly I:C). Following persistent antigen stimulation, the mice were treated with IL-2 and the therapeutic effects were analyzed. The results showed that compared with the mice that received transient antigen stimulation (boost twice), persistent antigen stimulation (boost more than 10 times) resulted in decrease of antigen specific IFN-γ and IL-2 production, reduction of memory CD8+ T cells, over-expression of immune checkpoint programmed cell death protein 1 (PD-1), and impaired the protective immunity against bacterial challenge. Treating the T cell functionally exhausted mice with IL-2 restored antigen-specific T cell responses and protective efficacy. In conclusion, persistent stimulation with M. tuberculosis antigens induced T cell dysfunction, which could be restored by complement of IL-2.
Collapse
Affiliation(s)
- Xun Liu
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,School of Basic Medical Sciences, Institute of Pathogen Biology, Lanzhou University, Lanzhou, China
| | - Fei Li
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,School of Basic Medical Sciences, Institute of Pathogen Biology, Lanzhou University, Lanzhou, China
| | - Hongxia Niu
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,School of Basic Medical Sciences, Institute of Pathogen Biology, Lanzhou University, Lanzhou, China
| | - Lan Ma
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,School of Basic Medical Sciences, Institute of Pathogen Biology, Lanzhou University, Lanzhou, China
| | - Jianzhu Chen
- Department of Biology, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Liang Peng
- Center of Life Science, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Chao Gan
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,School of Basic Medical Sciences, Institute of Pathogen Biology, Lanzhou University, Lanzhou, China
| | - Xingming Ma
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Bingdong Zhu
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,School of Basic Medical Sciences, Institute of Pathogen Biology, Lanzhou University, Lanzhou, China
| |
Collapse
|
115
|
Wang Y, Zhao N, Wu Z, Pan N, Shen X, Liu T, Wei F, You J, Xu W, Ren X. New insight on the correlation of metabolic status on 18F-FDG PET/CT with immune marker expression in patients with non-small cell lung cancer. Eur J Nucl Med Mol Imaging 2019; 47:1127-1136. [PMID: 31502013 DOI: 10.1007/s00259-019-04500-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/21/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Metabolic information obtained through 18F-flurodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) is used to evaluate malignancy by calculating the glucose uptake rate, and these parameters play important roles in determining the prognosis of non-small cell lung cancer (NSCLC). The expression of immune-related markers in tumor tissue reflects the immune status in the tumor microenvironment. However, there is lack of reports on the association between metabolic variables and intra-tumor immune markers. Herein, we investigate the correlation between metabolic status on 18F-FDG PET/CT and intra-tumor immunomarkers' expression in NSCLC patients. METHODS From April 2008 to August 2014, 763 patients were enrolled in the analysis to investigate the role of maximum standardized uptake value (SUVmax) in lung cancer. One hundred twenty-two tumor specimens were analyzed by immunohistochemistry (IHC) to intra-tumor immune cells and programmed death protein ligand 1(PD-L1) expression on tumor cells. The correlation between metabolic variables and the expression of tissue immune markers were analyzed. RESULTS SUVmax values have significant variations in different epidermal growth factor receptor (EGFR) statuses (wild type vs mutant type), high/low neutrophil-to-lymphocyte ratio (NLR) groups, and high/low platelets-to-lymphocyte ratio (PLR) groups (p < 0.001, p < 0.001, p = 0.003, respectively). SUVmax was an independent prognostic factor in lung cancer patients (p = 0.013). IHC demonstrated a statistically significant correlation between SUVmax and the expression of CD8 tumor-infiltrating lymphocytes (p = 0.015), CD163 tumor-associated macrophages (TAMs) (p = 0.003), and Foxp3-regulatory T cells (Tregs) (p = 0.004), as well as PD-1 and PD-L1 (p = 0.003 and p = 0.012, respectively). With respect to patient outcomes, disease stage, BMI, SUVmax, metabolic tumor volume (MTV), TLG (tumor lesion glycolysis), CD163-TAMs, CD11c-dendritic cells (DCs), PD-L1, and Tregs showed a statistically significant correlation with progression-free survival (PFS) (p < 0.001, 0.023, < 0.001, 0.007, 0.005, 0.004, 0.008, 0.048, and 0.014, respectively), and disease stage, SUVmax, MTV, TLG, CD163-TAMs, CD11c-DCs, and PD-L1 showed a statistically significant correlation with overall survival (OS) (p < 0.001, < 0.001, 0.014, 0.012, < 0.001, 0.001, and < 0.001, respectively). CONCLUSION This study revealed an association between metabolic variable and immune cell expression in the tumor microenvironment and suggests that SUVmax on 18F-FDG PET/CT could be a potential predictor for selecting candidates for immunotherapy.
Collapse
Affiliation(s)
- Yang Wang
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.,National Clinical Research Center of Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China
| | - Ning Zhao
- National Clinical Research Center of Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China.,Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Zhanbo Wu
- National Clinical Research Center of Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China.,Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Na Pan
- National Clinical Research Center of Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China.,Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Xuejie Shen
- National Clinical Research Center of Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China.,Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Ting Liu
- National Clinical Research Center of Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China.,Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Feng Wei
- National Clinical Research Center of Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China.,Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Jian You
- National Clinical Research Center of Cancer, Tianjin, 300060, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China. .,Department of Thoracic surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| | - Wengui Xu
- National Clinical Research Center of Cancer, Tianjin, 300060, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China. .,Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| | - Xiubao Ren
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China. .,National Clinical Research Center of Cancer, Tianjin, 300060, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China. .,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China.
| |
Collapse
|
116
|
Barham MS, Abrahams DA, Khayumbi J, Ongalo J, Tonui J, Campbell A, de Kock M, Ouma SG, Odhiambo FH, Hanekom WA, Gandhi NR, Day CL. HIV Infection Is Associated With Downregulation of BTLA Expression on Mycobacterium tuberculosis-Specific CD4 T Cells in Active Tuberculosis Disease. Front Immunol 2019; 10:1983. [PMID: 31497018 PMCID: PMC6712065 DOI: 10.3389/fimmu.2019.01983] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/06/2019] [Indexed: 12/20/2022] Open
Abstract
Nearly a quarter of the global population is infected with Mycobacterium tuberculosis (Mtb), with 10 million people developing active tuberculosis (TB) annually. Co-infection with human immunodeficiency virus (HIV) has long been recognized as a significant risk factor for progression to TB disease, yet the mechanisms whereby HIV impairs T cell-mediated control of Mtb infection remain poorly defined. We hypothesized that HIV infection may promote upregulation of inhibitory receptors on Mtb-specific CD4 T cells, a mechanism that has been associated with antigen-specific T cell dysfunction in chronic infections. Using cohorts of HIV-infected and HIV-uninfected individuals with latent Mtb infection (LTBI) and with active TB disease, we stimulated peripheral blood mononuclear cells (PBMC) for 6 hours with Mtb peptide pools and evaluated co-expression profiles of the inhibitory receptors BTLA, CTLA-4, and PD-1 on IFN-γ+/TNF-α+ Mtb-specific CD4 T cells. Mtb-specific CD4 T cells in all participant groups expressed predominately either one or no inhibitory receptors, unlike cytomegalovirus- and HIV-specific CD4 T cells circulating in the same individuals, which were predominately CTLA-4+PD-1+. There were no significant differences in inhibitory receptor expression profiles of Mtb-specific CD4 T cells between HIV-uninfected and HIV-infected individuals with LTBI. Surprisingly, BTLA expression, both alone and in combination with CTLA-4 and PD-1, was markedly downregulated on Mtb-specific CD4 T cells in HIV-infected individuals with active TB. Together, these data provide novel evidence that the majority of Mtb-specific CD4 T cells do not co-express multiple inhibitory receptors, regardless of HIV infection status; moreover, they highlight a previously unrecognized role of BTLA expression on Mtb-specific CD4 T cells that could be further explored as a potential biomarker of Mtb infection status, particularly in people living with HIV, the population at greatest risk for development of active TB disease.
Collapse
Affiliation(s)
- Morgan S Barham
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Deborah A Abrahams
- South African Tuberculosis Vaccine Initiative, School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Jeremiah Khayumbi
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Joshua Ongalo
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Joan Tonui
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Angela Campbell
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Marwou de Kock
- South African Tuberculosis Vaccine Initiative, School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Samuel Gurrion Ouma
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | | | - Willem A Hanekom
- South African Tuberculosis Vaccine Initiative, School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Neel R Gandhi
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Cheryl L Day
- Emory Vaccine Center, Emory University, Atlanta, GA, United States.,Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
117
|
Chen Y, Wang J, Wang X, Li X, Song J, Fang J, Liu X, Liu T, Wang D, Li Q, Wen S, Ma D, Xia J, Luo L, Zheng SG, Cui J, Zeng G, Chen L, Cheng B, Wang Z. Pik3ip1 Is a Negative Immune Regulator that Inhibits Antitumor T-Cell Immunity. Clin Cancer Res 2019; 25:6180-6194. [PMID: 31350312 DOI: 10.1158/1078-0432.ccr-18-4134] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 04/19/2019] [Accepted: 07/24/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Multiple negative regulators restrict the ability of T cells to attack tumors. This work demonstrates the role of PI3K-interacting protein 1 (Pik3ip1) in restraining T-cell responses and antitumor immunity. EXPERIMENTAL DESIGN An anti-Pik3ip1 mAb was generated to identify the Pik3ip1 expression pattern of hematopoietic cells. Pik3ip1 -/- mice and a Pik3ip1 fusion protein were generated to investigate the effect of Pik3ip1 on T-cell-mediated antitumor immunity in MC38 and B16-F10 tumor models. Immunoblotting and confocal microscopy were used to identify inhibitory effects of Pik3ip1 on T-cell receptor (TCR) signaling. Pik3ip1 expression was quantified, and its impact on T-cell function in human tumors was measured. RESULTS We demonstrated that Pik3ip1 was predominantly expressed on T cells and served as an essential rheostat for T-cell-mediated immunity. A Pik3ip1 genetic deficiency led to enhanced T-cell responsiveness upon immunization with a neoantigen. Pik3ip1 -/- mice exhibited a marked increase in antitumor immunity and were resistant to tumor growth. Furthermore, Pik3ip1 extracellular domain fusion protein enhanced MC38 tumor growth was observed. Mechanistically, we found that Pik3ip1 inhibited TCR signaling by mediating the degradation of SLP76 through Pik3ip1 oligomerization via its extracellular region. Consistent with the results from the mouse models, PIK3IP1 expression correlated with T-cell dysfunction in human tumors. CONCLUSIONS Our data reveal a critical role for Pik3ip1 as a novel inhibitory immune regulator of T-cell responses and provide a potential molecular target for cancer immunotherapy.
Collapse
Affiliation(s)
- Yichen Chen
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Jun Wang
- Department of Immunobiology and Yale Cancer Center, Yale University, New Haven, Connecticut
| | - Xi Wang
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Xinye Li
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Jingjing Song
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Juan Fang
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Xiangqi Liu
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Tao Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Dikan Wang
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Qunxing Li
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Shuqiong Wen
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Da Ma
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Juan Xia
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Liqun Luo
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, P.R. China
| | - Song Guo Zheng
- Department of Internal Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Gucheng Zeng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, P.R. China
| | - Lieping Chen
- Department of Immunobiology and Yale Cancer Center, Yale University, New Haven, Connecticut
| | - Bin Cheng
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China.
| | - Zhi Wang
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, P.R. China.
| |
Collapse
|
118
|
Duffy FJ, Thompson EG, Scriba TJ, Zak DE. Multinomial modelling of TB/HIV co-infection yields a robust predictive signature and generates hypotheses about the HIV+TB+ disease state. PLoS One 2019; 14:e0219322. [PMID: 31306460 PMCID: PMC6629068 DOI: 10.1371/journal.pone.0219322] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 06/20/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Current diagnostics are inadequate to meet the challenges presented by co-infection with Mycobacterium tuberculosis (Mtb) and HIV, the leading cause of death for HIV-infected individuals. Improved characterization of Mtb/HIV coinfection as a distinct disease state may lead to better identification and treatment of affected individuals. METHODS Four previously-published TB and HIV co-infection related datasets were used to train and validate multinomial machine learning classifiers that simultaneously predict TB and HIV status. Classifier predictive performance was measured using leave-one-out cross validation on the training set and blind predictive performance on multiple test sets using area under the ROC curve (AUC) as the performance metric. Linear modelling of signature gene expression was applied to systematically classify genes as TB-only, HIV-only or combined TB/HIV. RESULTS The optimal signature discovered was a 10-gene random forest multinomial signature that robustly discriminated active tuberculosis (TB) from other non-TB disease states with improved performance compared with previously published signatures (AUC: 0.87), and specifically discriminated active TB/HIV co-infection from all other conditions (AUC: 0.88). Signature genes exhibited a variety of transcriptional patterns including both TB-only and HIV-only response genes and genes with expression patterns driven by interactions between HIV and TB infection states, including the CD8+ T-cell receptor LAG3 and the apoptosis-related gene CERKL. CONCLUSIONS By explicitly including distinct disease states within the machine learning analysis framework, we developed a compact and highly diagnostic signature that simultaneously discriminates multiple disease states associated with Mtb/HIV co-infection. Examination of the expression patterns of signature genes suggests mechanisms underlying the unique inflammatory conditions associated with active TB in the presence of HIV. In particular, we observed that dysregulation of CD8+ effector T-cell and NK-cell associated genes may be an important feature of Mtb/HIV co-infection.
Collapse
Affiliation(s)
- Fergal J. Duffy
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States of America
- Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, WA, United States of America
| | - Ethan G. Thompson
- Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, WA, United States of America
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine & Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Daniel E. Zak
- Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, WA, United States of America
| |
Collapse
|
119
|
Li F, Liu X, Niu H, Lv W, Han X, Zhang Y, Zhu B. Persistent stimulation with Mycobacterium tuberculosis antigen impairs the proliferation and transcriptional program of hematopoietic cells in bone marrow. Mol Immunol 2019; 112:115-122. [PMID: 31082645 DOI: 10.1016/j.molimm.2019.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 05/01/2019] [Accepted: 05/04/2019] [Indexed: 02/07/2023]
Abstract
Mycobacterium tuberculosis (M. tuberculosis) persistent infection might cause the dysfunction of hematopoiesis. To investigate whether M. tuberculosis persistent antigen stimulation impairs the proliferation and differentiation of hematopoietic stem and progenitor cells characterized as lineage- c-Kit+ (LK cells), C57BL/6 mice were primed with Mycobacterium bovis Bacillus Calmette-Guérin (BCG) and boosted with a cocktail of M. tuberculosis antigens ESAT6, CFP10 and Mtb10.4-HspX (MH) along with adjuvant N, N'-dimethyl-N, N'-dioctadecylammonium bromide (DDA) plus polyinosinic-polycytidylic acid (Poly I:C) weekly for 12 or 22 weeks. The cytokine production by splenic T cells, proliferation of LK cells and transcriptional events during differentiation of bone marrow (BM) c-Kit+ cells were investigated. Meanwhile, the mice were treated with interleukin 2 (IL-2) and the therapeutic effects were analyzed. We found that antigen specific interferon-γ (IFN-γ) production by splenic CD4+ T cells increased following antigen stimulation for 12 weeks, but it declined after continuous stimulation for 22 weeks. The long-term exposure of mice to M. tuberculosis antigen compromised the proliferation of LK cells. Moreover, the expression of transcription factors in the c-Kit+ cells was adjusted, with up-regulation of IRF8 and Batf2 involved in myeloid differentiation and down-regulation of NOTCH1 and GATA2 participated in T-cell lineage commitment. The concentrations of IFN-γ in BM of the persistent antigen group were higher than that in sham control at the 12th week, while the concentrations of IL-2 in BM of the persistent antigen group were lower compared with the transient antigen stimulation control. Following IL-2 treatment, the concentrations of IL-2 in BM increased while IFN-γ got declined. IL-2 treatment could restore the expression levels of those transcription factors and the proliferating activity of LK cells impaired by persistent antigen stimulation. Our results indicate that M. tuberculosis antigen persistent stimulation decreases the proliferating activity of LK cells, promotes myelopoietic differentiation, and represses lymphopoietic differentiation as a consequence of elevated IFN-γ production. IL-2 supplementation contributes to maintaining the homeostasis of hemopoiesis.
Collapse
Affiliation(s)
- Fei Li
- Gansu Key Lab of Evidence Based Medicine and Clinical Transfer Medicine & Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, China.
| | - Xun Liu
- Gansu Key Lab of Evidence Based Medicine and Clinical Transfer Medicine & Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, China.
| | - Hongxia Niu
- Gansu Key Lab of Evidence Based Medicine and Clinical Transfer Medicine & Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, China.
| | - Wei Lv
- Gansu Key Lab of Evidence Based Medicine and Clinical Transfer Medicine & Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, China.
| | - Xue Han
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, China; Gansu Provincial Hospital, 204 West Donggang Road, Lanzhou 730000, China.
| | - Yifan Zhang
- Gansu Key Lab of Evidence Based Medicine and Clinical Transfer Medicine & Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, China.
| | - Bingdong Zhu
- Gansu Key Lab of Evidence Based Medicine and Clinical Transfer Medicine & Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, China.
| |
Collapse
|
120
|
Huang SF, Yang YY, Chou KT, Fung CP, Wang FD, Su WJ. Systemic proinflammation after Mycobacterium tuberculosis infection was correlated to the gut microbiome in HIV-uninfected humans. Eur J Clin Invest 2019; 49:e13068. [PMID: 30620398 DOI: 10.1111/eci.13068] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 01/01/2019] [Accepted: 01/06/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND The dysbiosis of gut microbiome and interaction with host immunity after Mycobacterium tuberculosis (MTB) infection are under investigation. We had found fatigue symptom concurrent with dysbiosis by decreasing the ratio of Firmicutes to Bacteroidetes (F/B ratio) in active tuberculosis (TB). The study aims to assess the inflammatory biomarkers and their interaction with gut microbiome in active TB and latent TB infection before starting anti-TB regimens. MATERIALS AND METHOD Interleukin-1 beta (IL-1B), IL-4, IL-6, IL-10, CD3+, CD4+, CD8+ T cells and interferon-gamma (IFN-γ) releasing assay (IGRA) were measured in 25 active TB patients, 32 LTBI subjects and 23 healthy controls (HC). Gut microbiome profiles were obtained using 16S rRNA MiSeq sequencing method. RESULTS The leucocytosis (7032 ± 387 cell/cum, P < 0.05), increase in IL-6 (229.7 ± 104 µg/dL, P < 0.05), and decrease in IL-4 (0.27 µg/dL ± 0.1, P < 0.05) were presented in active TB. The proportion of polymorphic neutrophil (PMN) in peripheral blood was positively related to the relative abundance of Bacteroidetes in LTBI and active TB (R2 = 0.23, P < 0.05). The F/B ratio was positively related to the detectable IL-1B in TB (R2 = 0.97, P < 0.01) and to the IL-4 in LTBI (R2 = 0.27, P < 0.05). In LTBI, the relative abundances of Coriobacteriaceae were positively related to the secretion of IFN-gamma against MTB-antigens more likely associated with of CD4+ T cell (R2 = 0.42, P < 0.05). CONCLUSION In active TB, dysbiosis with higher relative abundances of Bacteroidetes in stool and low F/B ratio was related to systemic proinflammation. In LTBI, dose-response relationship between peripheral PMN and relative abundances of Bacteroidetes was remained but not leads to systemic inflammation.
Collapse
Affiliation(s)
- Shiang-Fen Huang
- Institution of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Division of Infectious Disease, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C.,Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Ying-Ying Yang
- Institution of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C.,Institute of Public Health, Department of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan, R.O.C
| | - Kun-Ta Chou
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Chang-Phone Fung
- Institution of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Division of Infectious Disease, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Fu-Der Wang
- Division of Infectious Disease, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C.,School of Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Wei-Juin Su
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| |
Collapse
|
121
|
Intracellular Pathogens: Host Immunity and Microbial Persistence Strategies. J Immunol Res 2019; 2019:1356540. [PMID: 31111075 PMCID: PMC6487120 DOI: 10.1155/2019/1356540] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 01/18/2023] Open
Abstract
Infectious diseases caused by pathogens including viruses, bacteria, fungi, and parasites are ranked as the second leading cause of death worldwide by the World Health Organization. Despite tremendous improvements in global public health since 1950, a number of challenges remain to either prevent or eradicate infectious diseases. Many pathogens can cause acute infections that are effectively cleared by the host immunity, but a subcategory of these pathogens called "intracellular pathogens" can establish persistent and sometimes lifelong infections. Several of these intracellular pathogens manage to evade the host immune monitoring and cause disease by replicating inside the host cells. These pathogens have evolved diverse immune escape strategies and overcome immune responses by residing and multiplying inside host immune cells, primarily macrophages. While these intracellular pathogens that cause persistent infections are phylogenetically diverse and engage in diverse immune evasion and persistence strategies, they share common pathogen type-specific mechanisms during host-pathogen interaction inside host cells. Likewise, the host immune system is also equipped with a diverse range of effector functions to fight against the establishment of pathogen persistence and subsequent host damage. This article provides an overview of the immune effector functions used by the host to counter pathogens and various persistence strategies used by intracellular pathogens to counter host immunity, which enables their extended period of colonization in the host. The improved understanding of persistent intracellular pathogen-derived infections will contribute to develop improved disease diagnostics, therapeutics, and prophylactics.
Collapse
|
122
|
Ernst JD. Mechanisms of M. tuberculosis Immune Evasion as Challenges to TB Vaccine Design. Cell Host Microbe 2019; 24:34-42. [PMID: 30001523 DOI: 10.1016/j.chom.2018.06.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tuberculosis (TB) is a large global health problem, in part because of the long period of coevolution of the pathogen, Mycobacterium tuberculosis, and its human host. A major factor that sustains the global epidemic of TB is the lack of a sufficiently effective vaccine. While basic mechanisms of immunity that protect against TB have been identified, attempts to improve immunity to TB by vaccination have been disappointing. This Review discusses the mechanisms used by M. tuberculosis to evade innate and adaptive immunity and that likely limit the efficacy of vaccines developed to date. Despite multiple mechanisms of immune evasion, recent trials have indicated that effective TB vaccines remain an attainable goal. This Review discusses how knowledge from other systems can inform improvements on current vaccine approaches.
Collapse
Affiliation(s)
- Joel D Ernst
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
123
|
Ashwin H, Seifert K, Forrester S, Brown N, MacDonald S, James S, Lagos D, Timmis J, Mottram JC, Croft SL, Kaye PM. Tissue and host species-specific transcriptional changes in models of experimental visceral leishmaniasis. Wellcome Open Res 2019; 3:135. [PMID: 30542664 PMCID: PMC6248268 DOI: 10.12688/wellcomeopenres.14867.2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2018] [Indexed: 12/19/2022] Open
Abstract
Background: Human visceral leishmaniasis, caused by infection with Leishmania donovani or L. infantum, is a potentially fatal disease affecting 50,000-90,000 people yearly in 75 disease endemic countries, with more than 20,000 deaths reported. Experimental models of infection play a major role in understanding parasite biology, host-pathogen interaction, disease pathogenesis, and parasite transmission. In addition, they have an essential role in the identification and pre-clinical evaluation of new drugs and vaccines. However, our understanding of these models remains fragmentary. Although the immune response to Leishmania donovani infection in mice has been extensively characterized, transcriptomic analysis capturing the tissue-specific evolution of disease has yet to be reported. Methods: We provide an analysis of the transcriptome of spleen, liver and peripheral blood of BALB/c mice infected with L. donovani. Where possible, we compare our data in murine experimental visceral leishmaniasis with transcriptomic data in the public domain obtained from the study of L. donovani-infected hamsters and patients with human visceral leishmaniasis. Digitised whole slide images showing the histopathology in spleen and liver are made available via a dedicated website, www.leishpathnet.org. Results: Our analysis confirms marked tissue-specific alterations in the transcriptome of infected mice over time and identifies previously unrecognized parallels and differences between murine, hamster and human responses to infection. We show commonality of interferon-regulated genes whilst confirming a greater activation of type 2 immune pathways in infected hamsters compared to mice. Cytokine genes and genes encoding immune checkpoints were markedly tissue specific and dynamic in their expression, and pathways focused on non-immune cells reflected tissue specific immunopathology. Our data also addresses the value of measuring peripheral blood transcriptomics as a potential window into underlying systemic disease. Conclusions: Our transcriptomic data, coupled with histopathologic analysis of the tissue response, provide an additional resource to underpin future mechanistic studies and to guide clinical research.
Collapse
Affiliation(s)
- Helen Ashwin
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Karin Seifert
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Sarah Forrester
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Najmeeyah Brown
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Sandy MacDonald
- Bioscience Technology Facility, Deptartment of Biology, University of York, York, YO10 5DD, UK
| | - Sally James
- Bioscience Technology Facility, Deptartment of Biology, University of York, York, YO10 5DD, UK
| | - Dimitris Lagos
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Jon Timmis
- Dept of Electronic Engineering, University of York, York, YO10 5DD, UK
| | - Jeremy C Mottram
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Simon L. Croft
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Paul M. Kaye
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| |
Collapse
|
124
|
Liu YY, Sha W, Xu S, Gui XW, Xia L, Ji P, Wang S, Zhao GP, Zhang X, Chen Y, Wang Y. Identification of HLA-A2-Restricted Mycobacterial Lipoprotein Z Peptides Recognized by T CellsFrom Patients With ActiveTuberculosis Infection. Front Microbiol 2018; 9:3131. [PMID: 30622521 PMCID: PMC6308912 DOI: 10.3389/fmicb.2018.03131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 12/04/2018] [Indexed: 12/14/2022] Open
Abstract
Identification of HLA-restricted peptides derived from mycobacterial antigens that are endowed with high affinity and strong antigenicity is not only of interest in tuberculosis (TB) diagnostics and treatment efficacy evaluation, but might also provide potential candidates for the development of therapeutic vaccines against drug-resistant TB. Our previous work demonstrated that lipoprotein Z (LppZ) displayed high immunogenicity and antigenicity in active TB patients. In the present study, ten HLA-A2-restricted LppZ peptides (LppZp1-10) were predicted by bioinformatics, among which LppZp7 and LppZp10 were verified to possess high affinity to HLA-A2 molecules using T2 cell-based affinity binding assay. Moreover, results from ELISpot assay showed that both LppZp7 and LppZp10 peptides were able to induce more IFN-γ producing cells upon ex vivo stimulation of PBMC from HLA-A2+ active TB (ATB) patients as compared to those from healthy controls (HCs). Also, the numbers of LppZp7 and LppZp10-specific IFN-γ producing cells exhibited positive correlations with those of ESAT-6 peptide (E6p) or CFP-10 peptide (C10p) in ATB. Interestingly, stimulation with LppZp7/p10 mixture was able to induce higher intracellular expression of IFN-γ and IL-2 cytokines in CD8+ and CD4+ T cells from ATB as compared to HC, associated with lower expression of TNF-α in both CD8+ and CD4+ T cells. Taken together, HLA-A2-restricted LppZp7 and LppZp10 peptides display high immunoreactivity in HLA-matched ATB patients demonstrated by high responsiveness in both CD8+ and CD4+ T cells. With the ability to induce strong antigen-specific cellular responses, LppZp7 and LppZp10 are of potential value for the future applications in the prevention and control of TB.
Collapse
Affiliation(s)
- Yuan-Yong Liu
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, China.,Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Sha
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shiqiang Xu
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu-Wei Gui
- Clinic and Research Center of Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liliang Xia
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Ji
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujun Wang
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guo-Ping Zhao
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Xiao Zhang
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, China
| | - Yingying Chen
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| |
Collapse
|
125
|
Ashwin H, Seifert K, Forrester S, Brown N, MacDonald S, James S, Lagos D, Timmis J, Mottram JC, Croft SL, Kaye PM. Tissue and host species-specific transcriptional changes in models of experimental visceral leishmaniasis. Wellcome Open Res 2018; 3:135. [PMID: 30542664 PMCID: PMC6248268 DOI: 10.12688/wellcomeopenres.14867.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2018] [Indexed: 11/08/2023] Open
Abstract
Background: Human visceral leishmaniasis, caused by infection with Leishmania donovani or L. infantum, is a potentially fatal disease affecting 50,000-90,000 people yearly in 75 disease endemic countries, with more than 20,000 deaths reported. Experimental models of infection play a major role in understanding parasite biology, host-pathogen interaction, disease pathogenesis, and parasite transmission. In addition, they have an essential role in the identification and pre-clinical evaluation of new drugs and vaccines. However, our understanding of these models remains fragmentary. Although the immune response to Leishmania donovani infection in mice has been extensively characterized, transcriptomic analysis capturing the tissue-specific evolution of disease has yet to be reported. Methods: We provide an analysis of the transcriptome of spleen, liver and peripheral blood of BALB/c mice infected with L. donovani. Where possible, we compare our data in murine experimental visceral leishmaniasis with transcriptomic data in the public domain obtained from the study of L. donovani-infected hamsters and patients with human visceral leishmaniasis. Digitised whole slide images showing the histopathology in spleen and liver are made available via a dedicated website, www.leishpathnet.org. Results: Our analysis confirms marked tissue-specific alterations in the transcriptome of infected mice over time and identifies previously unrecognized parallels and differences between murine, hamster and human responses to infection. We show commonality of interferon-regulated genes whilst confirming a greater activation of type 2 immune pathways in infected hamsters compared to mice. Cytokine genes and genes encoding immune checkpoints were markedly tissue specific and dynamic in their expression, and pathways focused on non-immune cells reflected tissue specific immunopathology. Our data also addresses the value of measuring peripheral blood transcriptomics as a potential window into underlying systemic disease. Conclusions: Our transcriptomic data, coupled with histopathologic analysis of the tissue response, provide an additional resource to underpin future mechanistic studies and to guide clinical research.
Collapse
Affiliation(s)
- Helen Ashwin
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Karin Seifert
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Sarah Forrester
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Najmeeyah Brown
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Sandy MacDonald
- Bioscience Technology Facility, Deptartment of Biology, University of York, York, YO10 5DD, UK
| | - Sally James
- Bioscience Technology Facility, Deptartment of Biology, University of York, York, YO10 5DD, UK
| | - Dimitris Lagos
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Jon Timmis
- Dept of Electronic Engineering, University of York, York, YO10 5DD, UK
| | - Jeremy C Mottram
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Simon L. Croft
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Paul M. Kaye
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| |
Collapse
|
126
|
Olive AJ, Sassetti CM. Tolerating the Unwelcome Guest; How the Host Withstands Persistent Mycobacterium tuberculosis. Front Immunol 2018; 9:2094. [PMID: 30258448 PMCID: PMC6143787 DOI: 10.3389/fimmu.2018.02094] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 08/24/2018] [Indexed: 12/20/2022] Open
Abstract
Our understanding of the host response to infections has historically focused on “resistance” mechanisms that directly control pathogen replication. However, both pathogen effectors and antimicrobial immune pathways have the capacity to damage host tissue, and the ability to tolerate these insults can also be critical for host survival. These “tolerance” mechanisms may be equally as important as resistance to prevent disease in the context of a persistent infection, such as tuberculosis, when resistance mechanisms are ineffective and the pathogen persists in the tissue for long periods. Host tolerance encompasses a wide range of strategies, many of which involve regulation of the inflammatory response. Here we will examine general strategies used by macrophages and T cells to promote tolerance in the context of tuberculosis, and focus on pathways, such as regulation of inflammasome activation, that are emerging as common mediators of tolerance.
Collapse
Affiliation(s)
- Andrew J Olive
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Christopher M Sassetti
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
127
|
The exhausted CD4+CXCR5+ T cells involve the pathogenesis of human tuberculosis disease. Int J Infect Dis 2018; 74:1-9. [DOI: 10.1016/j.ijid.2018.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/31/2018] [Accepted: 06/14/2018] [Indexed: 12/18/2022] Open
|
128
|
Low Levels of T Cell Exhaustion in Tuberculous Lung Granulomas. Infect Immun 2018; 86:IAI.00426-18. [PMID: 29891540 DOI: 10.1128/iai.00426-18] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/01/2018] [Indexed: 02/06/2023] Open
Abstract
The hallmarks of pulmonary Mycobacterium tuberculosis infection are lung granulomas. These organized structures are composed of host immune cells whose purpose is to contain or clear infection, creating a complex hub of immune and bacterial cell activity, as well as limiting pathology in the lungs. Yet, given cellular activity and the potential for frequent interactions between host immune cells and M. tuberculosis-infected cells, we observed a surprisingly low quantity of cytokine-producing T cells (<10% of granuloma T cells) in our recent study of M. tuberculosis infection within nonhuman primate (NHP) granulomas. Various mechanisms could limit T cell function, and one hypothesis is T cell exhaustion. T cell exhaustion is proposed to result from continual antigen stimulation, inducing them to enter a state characterized by low cytokine production, low proliferation, and expression of a series of inhibitory receptors, the most common being PD-1, LAG-3, and CTLA-4. In this work, we characterized the expression of inhibitory receptors on T cells and the functionality of these cells in tuberculosis (TB) lung granulomas. We then used these experimental data to calibrate and inform an agent-based computational model that captures environmental, cellular, and bacterial dynamics within granulomas in lungs during M. tuberculosis infection. Together, the results of the modeling and the experimental work suggest that T cell exhaustion alone is not responsible for the low quantity of M. tuberculosis-responsive T cells observed within TB granulomas and that the lack of exhaustion is likely an intrinsic property of granuloma structure.
Collapse
|
129
|
Olive AJ, Smith CM, Kiritsy MC, Sassetti CM. The Phagocyte Oxidase Controls Tolerance to Mycobacterium tuberculosis Infection. THE JOURNAL OF IMMUNOLOGY 2018; 201:1705-1716. [PMID: 30061198 DOI: 10.4049/jimmunol.1800202] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/11/2018] [Indexed: 01/16/2023]
Abstract
Protection from infectious disease relies on two distinct strategies: antimicrobial resistance directly inhibits pathogen growth, whereas infection tolerance protects from the negative impact of infection on host health. A single immune mediator can differentially contribute to these strategies in distinct contexts, confounding our understanding of protection to different pathogens. For example, the NADPH-dependent phagocyte oxidase (Phox) complex produces antimicrobial superoxide and protects from tuberculosis (TB) in humans. However, Phox-deficient mice display no sustained resistance defects to Mycobacterium tuberculosis, suggesting a more complicated role for NADPH Phox complex than strictly controlling bacterial growth. We examined the mechanisms by which Phox contributes to protection from TB and found that mice lacking the Cybb subunit of Phox suffered from a specific defect in tolerance, which was caused by unregulated Caspase-1 activation, IL-1β production, and neutrophil influx into the lung. These studies imply that a defect in tolerance alone is sufficient to compromise immunity to M. tuberculosis and highlight a central role for Phox and Caspase-1 in regulating TB disease progression.
Collapse
Affiliation(s)
- Andrew J Olive
- University of Massachusetts Medical School, Worcester, MA 01605
| | - Clare M Smith
- University of Massachusetts Medical School, Worcester, MA 01605
| | | | | |
Collapse
|
130
|
Abstract
Protective immunity in tuberculosis (TB) is subject of debate in the TB research community, as this is key to fully understand TB pathogenesis and to develop new promising tools for TB diagnosis and prognosis as well as a more efficient TB vaccine. IFN-γ producing CD4+ T cells are key in TB control, but may not be sufficient to provide protection. Additional subsets have been identified that contribute to protection such as multifunctional and cytolytic T-cell subsets, including classical and nonclassical T cells as well as novel innate immune cell subsets resulting from trained immunity. However, to define protective immune responses against TB, the complexity of balancing TB immunity also has to be considered. In this review, insights into effector cell immunity and how this is modulated by regulatory cells, associated comorbidities and the host microbiome, is discussed. We systematically map how different suppressive immune cell subsets may affect effector cell responses at the local site of infection. We also dissect how common comorbidities such as HIV, helminths and diabetes may bias protective TB immunity towards pathogenic and regulatory responses. Finally, also the composition and diversity of the microbiome in the lung and gut could affect host TB immunity. Understanding these various aspects of the immunological balance in the human host is fundamental to prevent TB infection and disease.
Collapse
Affiliation(s)
- Susanna Brighenti
- Karolinska Institutet, Department of Medicine, Center for Infectious Medicine (CIM), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Simone A. Joosten
- Leiden University Medical Center, Department of Infectious Diseases, Leiden, The Netherlands
| |
Collapse
|
131
|
Tiberi S, du Plessis N, Walzl G, Vjecha MJ, Rao M, Ntoumi F, Mfinanga S, Kapata N, Mwaba P, McHugh TD, Ippolito G, Migliori GB, Maeurer MJ, Zumla A. Tuberculosis: progress and advances in development of new drugs, treatment regimens, and host-directed therapies. THE LANCET. INFECTIOUS DISEASES 2018; 18:e183-e198. [PMID: 29580819 DOI: 10.1016/s1473-3099(18)30110-5] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/19/2017] [Accepted: 01/02/2018] [Indexed: 12/16/2022]
Abstract
Tuberculosis remains the world's leading cause of death from an infectious disease, responsible for an estimated 1 674 000 deaths annually. WHO estimated 600 000 cases of rifampicin-resistant tuberculosis in 2016-of which 490 000 were multidrug resistant (MDR), with less than 50% survival after receiving recommended treatment regimens. Concerted efforts of stakeholders, advocates, and researchers are advancing further development of shorter course, more effective, safer, and better tolerated treatment regimens. We review the developmental pipeline and landscape of new and repurposed tuberculosis drugs, treatment regimens, and host-directed therapies (HDTs) for drug-sensitive and drug-resistant tuberculosis. 14 candidate drugs for drug-susceptible, drug-resistant, and latent tuberculosis are in clinical stages of drug development; nine are novel in phase 1 and 2 trials, and three new drugs are in advanced stages of development for MDR tuberculosis. Specific updates are provided on clinical trials of bedaquiline, delamanid, pretomanid, and other licensed or repurposed drugs that are undergoing investigation, including trials aimed at shortening duration of tuberculosis treatment, improving treatment outcomes and patient adherence, and reducing toxic effects. Ongoing clinical trials for shortening tuberculosis treatment duration, improving treatment outcomes in MDR tuberculosis, and preventing disease in people with latent tuberculosis infection are reviewed. A range of HDTs and immune-based treatments are under investigation as adjunctive therapy for shortening duration of therapy, preventing permanent lung injury, and improving treatment outcomes of MDR tuberculosis. We discuss the HDT development pipeline, ongoing clinical trials, and translational research efforts for adjunct tuberculosis treatment.
Collapse
Affiliation(s)
- Simon Tiberi
- Division of Infection, Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Nelita du Plessis
- South African Department of Science and Technology, and National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- South African Department of Science and Technology, and National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | | | - Martin Rao
- Champalimaud Foundation, Lisbon, Portugal; Krankenhaus Nordwest, Frankfurt, Germany
| | - Francine Ntoumi
- Fondation Congolaise pour la Recherche Medicale, and Faculte des Sciences et Techniques, Universite M Ngouabi, Brazzaville, Republic of the Congo
| | - Sayoki Mfinanga
- National Institute for Medical Research, Muhimbili Medical Research Centre, Dar es Salaam, Tanzania
| | - Nathan Kapata
- Institute of Public Health, Ministry of Health, Lusaka, Zambia
| | - Peter Mwaba
- UNZA-UCLMS Research and Training Programme, and Apex University, Lusaka, Zambia
| | - Timothy D McHugh
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, UK
| | - Giuseppe Ippolito
- National Institute for Infectious Disease, L Spallanzani, Rome, Italy
| | - Giovanni Battista Migliori
- World Health Organization Collaborating Centre for Tuberculosis and Lung Diseases, Maugeri Care and Research Institute, Istituto di Ricovero e Cura a Carattere Sceintifico, Tradate, Italy
| | - Markus J Maeurer
- Champalimaud Foundation, Lisbon, Portugal; Krankenhaus Nordwest, Frankfurt, Germany
| | - Alimuddin Zumla
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, UK; National Institute of Health and Research Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK.
| |
Collapse
|
132
|
Abstract
T-cell immunoglobulin and mucin domain 3 (Tim-3) is a transmembrane protein that in both mice and humans has been shown to possess various functions in a context-dependent manner. Thus, Tim-3 has been associated with both inhibitory and co-stimulatory function, depending in part on the specific cell type and immune response course. Though originally described on T cells, Tim-3 is now known to be expressed by both lymphoid and non-lymphoid cells within the immune system and even by non-immune cells. In addition, though widely thought of as a negative regulator of immunity, Tim-3 has been shown in more recent studies to have a positive function on both myeloid and lymphoid cells, including T cells. Tim-3 is often expressed at a high level on exhausted T cells in tumors and chronic infection and may engage in crosstalk with other so-called "checkpoint" molecules such as PD-1. Thus, Tim-3 has emerged as a possible therapeutic target, which is being actively explored both pre-clinically and clinically. However, recent research suggests a more complex in vivo role for this protein, compared with other targets in this area.
Collapse
Affiliation(s)
- Hridesh Banerjee
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| |
Collapse
|
133
|
Hart P, Copland A, Diogo GR, Harris S, Spallek R, Oehlmann W, Singh M, Basile J, Rottenberg M, Paul MJ, Reljic R. Nanoparticle-Fusion Protein Complexes Protect against Mycobacterium tuberculosis Infection. Mol Ther 2018; 26:822-833. [PMID: 29518353 PMCID: PMC5910664 DOI: 10.1016/j.ymthe.2017.12.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 11/26/2022] Open
Abstract
Tuberculosis (TB) is the leading cause of death from infectious disease, and the current vaccine, Bacillus Calmette-Guerin (BCG), is inadequate. Nanoparticles (NPs) are an emerging vaccine technology, with recent successes in oncology and infectious diseases. NPs have been exploited as antigen delivery systems and also for their adjuvantic properties. However, the mechanisms underlying their immunological activity remain obscure. Here, we developed a novel mucosal TB vaccine (Nano-FP1) based upon yellow carnauba wax NPs (YC-NPs), coated with a fusion protein consisting of three Mycobacterium tuberculosis (Mtb) antigens: Acr, Ag85B, and HBHA. Mucosal immunization of BCG-primed mice with Nano-FP1 significantly enhanced protection in animals challenged with low-dose, aerosolized Mtb. Bacterial control by Nano-FP1 was associated with dramatically enhanced cellular immunity compared to BCG, including superior CD4+ and CD8+ T cell proliferation, tissue-resident memory T cell (Trm) seeding in the lungs, and cytokine polyfunctionality. Alongside these effects, we also observed potent humoral responses, such as the generation of Ag85B-specific serum IgG and respiratory IgA. Finally, we found that YC-NPs were able to activate antigen-presenting cells via an unconventional IRF-3-associated activation signature, without the production of potentially harmful inflammatory mediators, providing a mechanistic framework for vaccine efficacy and future development.
Collapse
Affiliation(s)
- Peter Hart
- St George's Medical School, University of London, London SW17 0RE, UK
| | - Alastair Copland
- St George's Medical School, University of London, London SW17 0RE, UK
| | | | - Shane Harris
- St George's Medical School, University of London, London SW17 0RE, UK
| | | | | | | | | | | | - Matthew John Paul
- St George's Medical School, University of London, London SW17 0RE, UK
| | - Rajko Reljic
- St George's Medical School, University of London, London SW17 0RE, UK.
| |
Collapse
|
134
|
Xia AL, Wang JC, Yang K, Ji D, Huang ZM, Xu Y. Genomic and epigenomic perspectives of T-cell exhaustion in cancer. Brief Funct Genomics 2018. [PMID: 29518177 DOI: 10.1093/bfgp/ely005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- An-Liang Xia
- Department of General Surgery, Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jin-Cheng Wang
- Department of General Surgery, Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kun Yang
- Department of Medical Engineering, 302 Hospital of PLA, Beijing, China
| | - Dong Ji
- Liver Cirrhosis Treatment and Research Center II, 302 Military Hospital of China 100 XiSiHuan Middle Road, FengTai District, Beijing, China
| | - Zheng-Ming Huang
- Department of Clinical Pharmacology, 302 Hospital of PLA, Beijing, China
| | - Yong Xu
- Department of Nephrology, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| |
Collapse
|
135
|
Abstract
Vaccination approaches eliciting strong CD4 T cell responses provide only weak protection from tuberculosis. In this issue of Cell Host & Microbe, Moguche et al. (2017) show that T cells specific for different immunodominant vaccine antigens can fail to protect for opposite reasons, highlighting a major challenge in tuberculosis vaccine design.
Collapse
Affiliation(s)
- Daniel L Barber
- T Lymphocyte Biology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA.
| |
Collapse
|
136
|
Moguche AO, Musvosvi M, Penn-Nicholson A, Plumlee CR, Mearns H, Geldenhuys H, Smit E, Abrahams D, Rozot V, Dintwe O, Hoff ST, Kromann I, Ruhwald M, Bang P, Larson RP, Shafiani S, Ma S, Sherman DR, Sette A, Lindestam Arlehamn CS, McKinney DM, Maecker H, Hanekom WA, Hatherill M, Andersen P, Scriba TJ, Urdahl KB. Antigen Availability Shapes T Cell Differentiation and Function during Tuberculosis. Cell Host Microbe 2018; 21:695-706.e5. [PMID: 28618268 DOI: 10.1016/j.chom.2017.05.012] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 04/03/2017] [Accepted: 05/30/2017] [Indexed: 01/20/2023]
Abstract
CD4 T cells are critical for protective immunity against Mycobacterium tuberculosis (Mtb), the cause of tuberculosis (TB). Yet to date, TB vaccine candidates that boost antigen-specific CD4 T cells have conferred little or no protection. Here we examined CD4 T cell responses to two leading TB vaccine antigens, ESAT-6 and Ag85B, in Mtb-infected mice and in vaccinated humans with and without underlying Mtb infection. In both species, Mtb infection drove ESAT-6-specific T cells to be more differentiated than Ag85B-specific T cells. The ability of each T cell population to control Mtb in the lungs of mice was restricted for opposite reasons: Ag85B-specific T cells were limited by reduced antigen expression during persistent infection, whereas ESAT-6-specific T cells became functionally exhausted due to chronic antigenic stimulation. Our findings suggest that different vaccination strategies will be required to optimize protection mediated by T cells recognizing antigens expressed at distinct stages of Mtb infection.
Collapse
Affiliation(s)
- Albanus O Moguche
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA; Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Munyaradzi Musvosvi
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Adam Penn-Nicholson
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | | | - Helen Mearns
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Hennie Geldenhuys
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Erica Smit
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Deborah Abrahams
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Virginie Rozot
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - One Dintwe
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Søren T Hoff
- Statens Serum Institut (SSI), 2300 Copenhagen, Denmark
| | | | | | - Peter Bang
- Statens Serum Institut (SSI), 2300 Copenhagen, Denmark
| | - Ryan P Larson
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA
| | - Shahin Shafiani
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA
| | - Shuyi Ma
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA
| | - David R Sherman
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA
| | - Alessandro Sette
- Department of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla 92037, USA
| | | | - Denise M McKinney
- Department of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla 92037, USA
| | - Holden Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Willem A Hanekom
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | | | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa.
| | - Kevin B Urdahl
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA; Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
137
|
Abstract
The upregulation of immune checkpoint molecules, such as programmed cell death protein 1 (PD1) and cytotoxic T lymphocyte antigen 4 (CTLA4), on immune cells occurs during acute infections, such as malaria, as well as during chronic persistent viral infections, including HIV and hepatitis B virus. These pathways are important for preventing immune-driven pathology but can also limit immune-mediated clearance of the infection. The recent success of immune checkpoint blockade in cancer therapy suggests that targeting these pathways would also be effective for preventing and treating a range of infectious diseases. Here, we review our current understanding of immune checkpoint pathways in the pathogenesis of infectious diseases and discuss the potential for therapeutically targeting these pathways in this setting.
Collapse
Affiliation(s)
- Michelle N Wykes
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, Queensland 4006, Australia
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3000, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria 3004, Australia
| |
Collapse
|
138
|
Li YM, Shi YY, Li Y, Yan L, Tang JT, Bai YJ, Wu XJ, Dai B, Zou YG, Wang LL. Soluble Tim-3 and Gal-9 are associated with renal allograft dysfunction in kidney transplant recipients: A cross-sectional study. Int Immunopharmacol 2018; 55:330-335. [PMID: 29310109 DOI: 10.1016/j.intimp.2018.01.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/22/2017] [Accepted: 01/04/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND T cell immunoglobulin mucin-3 (Tim-3) has been reported to participate in the regulation of immune response and the induction of allograft tolerance. However, the association between Tim-3 and renal allograft dysfunction is unclear. We studied the expression of cellular and soluble Tim-3 (sTim-3), soluble galectin-9 (sGal-9) and carcinoembryonic antigen-related cell adhesion molecule-1 (sCEACAM-1) in kidney transplantation recipients (KTRs) to explore their roles in allograft dysfunction. METHODS 96 KTRs (53 with stable graft and 43 with graft dysfunction) and 30 healthy controls (HC) were enrolled. Among the KTRs, 55 used Tacrolimus (TAC) and 41 used Sirolimus (SRL). In the dysfunction group, 29 recipients have undergone graft biopsy and 14 were classified as biopsy-proven rejection (BPR). Cellular Tim-3 was determined by flow cytometry. sTim-3 was determined by ELISA. sGal-9 and sCEACAM-1 were determined by Bio-Plex® suspension array system. RESULTS KTRs with renal dysfunction showed significantly higher levels of sTim-3 and sGal-9 but similar levels of cellular Tim-3 and sCEACAM-1 compared with stable recipients. Correlation analysis revealed that estimated glomerular filtration rate (eGFR) was negatively associated with sTim-3 and sGal-9. Both BPR and non-BPR groups showed comparable levels of Tim-3, Gal-9 and CEACAM-1. Moreover, SRL group showed significantly higher levels of sCEACAM-1 than TAC and HC groups. CONCLUSIONS sTim-3 and sGal-9 were promising biomarkers for allograft dysfunction, but unable to differentiate allograft rejection from other causes of renal dysfunction in KTRs. Moreover, long-term administration of sirolimus would up-regulate sCEACAM-1 level, while exert similar regulatory effects on Tim-3 and Gal-9 compared to tacrolimus.
Collapse
Affiliation(s)
- Ya Mei Li
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Ying Shi
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Li
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Yan
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jiang Tao Tang
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Juan Bai
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiao Juan Wu
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Dai
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Gao Zou
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Lan Wang
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
139
|
Zhu H, Gu Y, Xue Y, Yuan M, Cao X, Liu Q. CXCR2 + MDSCs promote breast cancer progression by inducing EMT and activated T cell exhaustion. Oncotarget 2017; 8:114554-114567. [PMID: 29383101 PMCID: PMC5777713 DOI: 10.18632/oncotarget.23020] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/14/2017] [Indexed: 12/02/2022] Open
Abstract
Although myeloid-derived suppressor cells (MDSCs) have been demonstrated to contribute to tumor initiation, progression and metastasis, however, which MDSC subsets are preferentially expanded and activated, and what's the key molecular mechanism responsible for specific MDSC subsets in promoting tumor progression need to be fully addressed. Here we identify that Ly6GmiLy6CloCD11b+CXCR2+ subpopulation (named CXCR2+ MDSCs) are predominately expanded and recruited in systemic and local tumor microenvironment during breast cancer progression and metastasis. The proportion of CXCR2+ MDSCs is inversely correlated with the infiltration of CD4+ or CD8+ T cells. Besides, CXCR2+ MDSCs promote breast cancer growth and metastasis to lung and/or lymph node in vivo. Furthermore, CXCR2+ MDSCs induce epithelial mesenchymal transition (EMT) of breast cancer cells via IL-6. Moreover, CXCR2+ MDSCs upregulate the expression of immunosuppressive molecules programmed cell death protein 1(PD1), PD1 ligand 1 (PDL1), lymphocyte activation gene 3 protein (LAG3), cytotoxic T lymphocyte antigen 4 (CTLA4), and T cell immunoglobulin domain and mucin domain protein 3 (TIM3) on CD4+ or CD8+ T cells, and induce exhaustion of the activated T cells partially via IFN-γ. These results demonstrate that CXCR2+ MDSCs accelerate breast cancer progression via directly inducing cancer cell EMT and indirectly promoting T cell exhaustion, suggesting that CXCR2+ MDSCs may be a potential therapeutic target of breast cancer.
Collapse
Affiliation(s)
- Ha Zhu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Yan Gu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Yiquan Xue
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Ming Yuan
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Qiuyan Liu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
140
|
Carpenter SM, Yang JD, Lee J, Barreira-Silva P, Behar SM. Vaccine-elicited memory CD4+ T cell expansion is impaired in the lungs during tuberculosis. PLoS Pathog 2017; 13:e1006704. [PMID: 29176787 PMCID: PMC5720822 DOI: 10.1371/journal.ppat.1006704] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 12/07/2017] [Accepted: 10/19/2017] [Indexed: 12/17/2022] Open
Abstract
Immunological memory is the key biological process that makes vaccines possible. Although tuberculosis vaccines elicit protective immunity in animals, few provide durable protection. To understand why protection is transient, we evaluated the ability of memory CD4+ T cells to expand, differentiate, and control Mycobacterium tuberculosis. Both naïve and memory CD4+ T cells initially proliferated exponentially, and the accumulation of memory T cells in the lung correlated with early bacterial control. However, later during infection, memory CD4+ T cell proliferation was curtailed and no protection was observed. We show that memory CD4+ T cells are first activated in the LN and their recruitment to the lung attenuates bacterial growth. However, their interaction with Mtb-infected macrophages does not promote continued proliferation. We conclude that a lack of sustained expansion by memory-derived T cells in the lung limits the durability of their protection, linking their slower expansion with transient protection in vaccinated mice. Vaccines elicit pathogen-specific memory T cells whose early and potent activation upon infection should provide long-lasting control of bacterial growth. Although many experimental vaccines generate memory CD4+ T cells and can control the growth of Mycobacterium tuberculosis (Mtb) early during infection, none reliably provide protection from pulmonary tuberculosis (TB) that is durable. Although the etiology of the clinical failure of memory T cells is not well understood, few studies monitor memory T cell fate and function throughout chronic infection. Using both clonal and polyclonal models of Mtb-specific memory CD4+ T cell function during TB, we show that the expansion of memory-derived T cell responses is impaired in the lungs, compared with the primary (naïve) CD4 response. Despite expressing a protective effector phenotype, and reducing bacterial growth early after Mtb challenge, we further show that memory CD4+ T cells do not proliferate in response to Mtb-infected macrophages. Their impaired expansion corresponded with waning protection in vaccinated mice later during infection. We propose that both the induction of memory T cell proliferation by infected macrophages, and the durability of vaccine-elicited T cell responses during TB should serve as preclinical vaccine benchmarks.
Collapse
Affiliation(s)
- Stephen M. Carpenter
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States of America
- Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
- * E-mail: (SMB); (SMC)
| | - Jason D. Yang
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Jinhee Lee
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Palmira Barreira-Silva
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Samuel M. Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (SMB); (SMC)
| |
Collapse
|
141
|
Li F, Dang J, Jiang M, He M, Yang M, Li J, Hao H, Zhou Y, Zuo W, Xie Y, Deng D. Upregulation of Tim-3 expression at feto-maternal interface may explain embryo survival in the CBAxDBA/2 model of abortion. Am J Reprod Immunol 2017; 79. [PMID: 29083087 DOI: 10.1111/aji.12775] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/02/2017] [Indexed: 01/08/2023] Open
Abstract
PROBLEM To understand the mechanisms of action of Tim-3 at the maternal-fetal interface and explore how Tim-3 might be involved in the pathogenesis of abortion by constructing an in vitro trophoblast-lymphocyte system. METHODS OF STUDY Female CBA/J × male DBA/2 matings were used as the abortion-prone model and CBA/J × male BALB/c matings as control. The expression of Tim-3 at the maternal-fetal interface and in the peripheral blood lymphocytes was measured by immunohistochemistry and Western blotting. The proliferation index of lymphocytes and levels of Th1/Th2-derived cytokines in peripheral blood and in the co-culture system were determined using CCK-8 assay and ELISA, respectively. RESULTS The expression level of Tim-3 was higher in abortion-prone matings than that of control (P < .05). A preponderance of Th1 was observed in the co-culture system in the abortion-prone mating group. Recombinant Tim-3 Ig reversed the imbalance of Th1/Th2 immunity of abortion-prone matings by suppressing the secretion of IFN-γ and IL-2 but had no direct effect on the generation of IL-4. CONCLUSION Tim-3 might contribute to successful pregnancy by restraining Th1 bias, and the maternal immune system might develop a strategy including upregulation of Tim-3 at the maternal-fetal interface and in peripheral blood so as to maintain moderate inflammatory responses against miscarriage.
Collapse
Affiliation(s)
- Fanfan Li
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Dang
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Jiang
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Gynecology and Obstetrics, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Jiangsu, China
| | - Mengzhou He
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meitao Yang
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Li
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Faculty of Reproductive Medical Center of the Third Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyan Hao
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuan Zhou
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Reproductive Medical Center, Tangdu Hospital, The Fourth Military Medical University, xi'an, China
| | - Wei Zuo
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Faculty of Department of Orthopedics, Pu Ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yin Xie
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dongrui Deng
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
142
|
Zhou Y, Du J, Hou HY, Lu YF, Yu J, Mao LY, Wang F, Sun ZY. Application of ImmunoScore Model for the Differentiation between Active Tuberculosis and Latent Tuberculosis Infection as Well as Monitoring Anti-tuberculosis Therapy. Front Cell Infect Microbiol 2017; 7:457. [PMID: 29164066 PMCID: PMC5670161 DOI: 10.3389/fcimb.2017.00457] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/12/2017] [Indexed: 01/17/2023] Open
Abstract
Tuberculosis (TB) is a leading global public health problem. To achieve the end TB strategy, non-invasive markers for diagnosis and treatment monitoring of TB disease are urgently needed, especially in high-endemic countries such as China. Interferon-gamma release assays (IGRAs) and tuberculin skin test (TST), frequently used immunological methods for TB detection, are intrinsically unable to discriminate active tuberculosis (ATB) from latent tuberculosis infection (LTBI). Thus, the specificity of these methods in the diagnosis of ATB is dependent upon the local prevalence of LTBI. The pathogen-detecting methods such as acid-fast staining and culture, all have limitations in clinical application. ImmunoScore (IS) is a new promising prognostic tool which was commonly used in tumor. However, the importance of host immunity has also been demonstrated in TB pathogenesis, which implies the possibility of using IS model for ATB diagnosis and therapy monitoring. In the present study, we focused on the performance of IS model in the differentiation between ATB and LTBI and in treatment monitoring of TB disease. We have totally screened five immunological markers (four non-specific markers and one TB-specific marker) and successfully established IS model by using Lasso logistic regression analysis. As expected, the IS model can effectively distinguish ATB from LTBI (with a sensitivity of 95.7% and a specificity of 92.1%) and also has potential value in the treatment monitoring of TB disease.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Du
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, China
| | - Hong-Yan Hou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-Fang Lu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Yan Mao
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zi-Yong Sun
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
143
|
Kaufmann SHE, Dorhoi A, Hotchkiss RS, Bartenschlager R. Host-directed therapies for bacterial and viral infections. Nat Rev Drug Discov 2017; 17:35-56. [PMID: 28935918 PMCID: PMC7097079 DOI: 10.1038/nrd.2017.162] [Citation(s) in RCA: 479] [Impact Index Per Article: 59.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Host-directed therapy (HDT) is a novel approach in the field of anti-infectives for overcoming antimicrobial resistance. HDT aims to interfere with host cell factors that are required by a pathogen for replication or persistence, to enhance protective immune responses against a pathogen, to reduce exacerbated inflammation and to balance immune reactivity at sites of pathology. HDTs encompassing the 'shock and kill' strategy or the delivery of recombinant interferons are possible approaches to treat HIV infections. HDTs that suppress the cytokine storm that is induced by some acute viral infections represent a promising concept. In tuberculosis, HDT aims to enhance the antimicrobial activities of phagocytes through phagosomal maturation, autophagy and antimicrobial peptides. HDTs also curtail inflammation through interference with soluble (such as eicosanoids or cytokines) or cellular (co-stimulatory molecules) factors and modulate granulomas to allow the access of antimicrobials or to restrict tissue damage. Numerous parallels between the immunological abnormalities that occur in sepsis and cancer indicate that the HDTs that are effective in oncology may also hold promise in sepsis. Advances in immune phenotyping, genetic screening and biosignatures will help to guide drug therapy to optimize the host response. Combinations of canonical pathogen-directed drugs and novel HDTs will become indispensable in treating emerging infections and diseases caused by drug-resistant pathogens.
Host-directed therapy (HDT) aims to interfere with host cell factors that are required by a pathogen for replication or persistence. In this Review, Kaufmannet al. describe recent progress in the development of HDTs for the treatment of viral and bacterial infections and the challenges in bringing these approaches to the clinic. Despite the recent increase in the development of antivirals and antibiotics, antimicrobial resistance and the lack of broad-spectrum virus-targeting drugs are still important issues and additional alternative approaches to treat infectious diseases are urgently needed. Host-directed therapy (HDT) is an emerging approach in the field of anti-infectives. The strategy behind HDT is to interfere with host cell factors that are required by a pathogen for replication or persistence, to enhance protective immune responses against a pathogen, to reduce exacerbated inflammation and to balance immune reactivity at sites of pathology. Although HDTs encompassing interferons are well established for the treatment of chronic viral hepatitis, novel strategies aimed at the functional cure of persistent viral infections and the development of broad-spectrum antivirals against emerging viruses seem to be crucial. In chronic bacterial infections, such as tuberculosis, HDT strategies aim to enhance the antimicrobial activities of phagocytes and to curtail inflammation through interference with soluble factors (such as eicosanoids and cytokines) or cellular factors (such as co-stimulatory molecules). This Review describes current progress in the development of HDTs for viral and bacterial infections, including sepsis, and the challenges in bringing these new approaches to the clinic.
Collapse
Affiliation(s)
- Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Anca Dorhoi
- Department of Immunology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany.,Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Richard S Hotchkiss
- Departments of Anesthesiology, Medicine, and Surgery, Washington University School of Medicine, St Louis, 660 S. Euclid, St Louis, Missouri 63110, USA
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany.,Division of Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
144
|
Amelio P, Portevin D, Reither K, Mhimbira F, Mpina M, Tumbo A, Nickel B, Marti H, Knopp S, Ding S, Penn-Nicholson A, Darboe F, Ohmiti K, Scriba TJ, Pantaleo G, Daubenberger C, Perreau M. Mixed Th1 and Th2 Mycobacterium tuberculosis-specific CD4 T cell responses in patients with active pulmonary tuberculosis from Tanzania. PLoS Negl Trop Dis 2017; 11:e0005817. [PMID: 28759590 PMCID: PMC5552332 DOI: 10.1371/journal.pntd.0005817] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/10/2017] [Accepted: 07/19/2017] [Indexed: 12/22/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) and helminth infections elicit antagonistic immune effector functions and are co-endemic in several regions of the world. We therefore hypothesized that helminth infection may influence Mtb-specific T-cell immune responses. We evaluated the cytokine profile of Mtb-specific T cells in 72 individuals with pulmonary TB disease recruited from two Sub-Saharan regions with high and moderate helminth burden i.e. 55 from Tanzania (TZ) and 17 from South Africa (SA), respectively. We showed that Mtb-specific CD4 T-cell functional profile of TB patients from Tanzania are primarily composed of polyfunctional Th1 and Th2 cells, associated with increased expression of Gata-3 and reduced expression of T-bet in memory CD4 T cells. In contrast, the cytokine profile of Mtb-specific CD4 T cells of TB patients from SA was dominated by single IFN-γ and dual IFN-γ/TNF-α and associated with TB-induced systemic inflammation and elevated serum levels of type I IFNs. Of note, the proportion of patients with Mtb-specific CD8 T cells was significantly reduced in Mtb/helminth co-infected patients from TZ. It is likely that the underlying helminth infection and possibly genetic and other unknown environmental factors may have caused the induction of mixed Th1/Th2 Mtb-specific CD4 T cell responses in patients from TZ. Taken together, these results indicate that the generation of Mtb-specific CD4 and CD8 T cell responses may be substantially influenced by environmental factors in vivo. These observations may have major impact in the identification of immune biomarkers of disease status and correlates of protection. Mycobacterium tuberculosis (Mtb) and helminth infections are co-endemic in several regions of the world and their immune responses may be mutually antagonistic. We therefore hypothesized that helminth infection would impact and potentially shape Mtb-specific T-cell responses and systemic inflammation in patients suffering from active pulmonary tuberculosis (TB) enrolled from two helminth endemic regions i.e. Tanzania (TZ) and South Africa (SA). In this study, we demonstrate for the first time that TB patients from SA and TZ harbor distinct immune responses to Mtb antigens. Indeed, we showed that Mtb-specific CD4 T-cell responses of TB patients from TZ were composed by a mixed T helper type 1 (Th1) and Th2 responses. In contrast, the cytokine profile of Mtb-specific CD4 T cells of TB patients from SA was dominated by Th1 cells and associated with TB-induced systemic inflammation and elevated serum levels of type I IFN. Taken together, these data indicate that Mtb-specific T-cell responses are diverse in human populations and can be strongly influenced by host and pathogen genetic background, co-infections and yet unknown environmental factors. Identification of correlates of risk and protection from TB disease will help in the rational development of protective T-cell based vaccines against TB, early monitoring TB treatment outcomes and focused follow up of high risk populations.
Collapse
Affiliation(s)
- Patrizia Amelio
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Damien Portevin
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Klaus Reither
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | | | | | - Beatrice Nickel
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Hanspeter Marti
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Stefanie Knopp
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Song Ding
- EuroVacc Foundation, Lausanne, Switzerland
| | - Adam Penn-Nicholson
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, South Africa
| | - Fatoumatta Darboe
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, South Africa
| | - Khalid Ohmiti
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, South Africa
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- SVRI, Lausanne, Switzerland
| | - Claudia Daubenberger
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
145
|
Das M, Zhu C, Kuchroo VK. Tim-3 and its role in regulating anti-tumor immunity. Immunol Rev 2017; 276:97-111. [PMID: 28258697 DOI: 10.1111/imr.12520] [Citation(s) in RCA: 617] [Impact Index Per Article: 77.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/19/2016] [Indexed: 12/13/2022]
Abstract
Immunotherapy is being increasingly recognized as a key therapeutic modality to treat cancer and represents one of the most exciting treatments for the disease. Fighting cancer with immunotherapy has revolutionized treatment for some patients and therapies targeting the immune checkpoint molecules such as CTLA-4 and PD-1 have achieved durable responses in melanoma, renal cancer, Hodgkin's diseases and lung cancer. However, the success rate of these treatments has been low and a large number of cancers, including colorectal cancer remain largely refractory to CTLA-4 and PD-1 blockade. This has provided impetus to identify other co-inhibitory receptors that could be exploited to enhance response rates of current immunotherapeutic agents and achieve responses to the cancers that are refectory to immunotherapy. Tim-3 is a co-inhibitory receptor that is expressed on IFN-g-producing T cells, FoxP3+ Treg cells and innate immune cells (macrophages and dendritic cells) where it has been shown to suppress their responses upon interaction with their ligand(s). Tim-3 has gained prominence as a potential candidate for cancer immunotherapy, where it has been shown that in vivo blockade of Tim-3 with other check-point inhibitors enhances anti-tumor immunity and suppresses tumor growth in several preclinical tumor models. This review discusses the recent findings on Tim-3, the role it plays in regulating immune responses in different cell types and the rationale for targeting Tim-3 for effective cancer immunotherapy.
Collapse
Affiliation(s)
- Madhumita Das
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.,Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Chen Zhu
- Discovery Biology, Research and Development, Sanofi US, Cambridge, MA, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.,Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
146
|
Mycobacterium tuberculosis Rv3615c is a highly immunodominant antigen and specifically induces potent Th1-type immune responses in tuberculosis pleurisy. Clin Sci (Lond) 2017; 131:1859-1876. [PMID: 28588103 DOI: 10.1042/cs20170205] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/04/2017] [Accepted: 06/06/2017] [Indexed: 02/05/2023]
Abstract
T-cell responses have been demonstrated to be essential for preventing Mycobacterium tuberculosis infection. The Th1-cytokines produced by T cells, such as INF-γ, IL-2, and TNF-α, not only limit the invasion of M. tuberculosis but also eliminate the pathogen at the site of infection. Bacillus Calmette-Guérin (BCG) is known to induce Th1-type responses but the protection is inadequate. Identification of immunogenic components, in addition to those expressed in BCG, and induction of a broad spectrum of Th1-type responses provide options for generating sufficient adaptive immunity. Here, we studied human pulmonary T-cell responses induced by the M. tuberculosis-specific antigen Rv3615c, a protein with a similar size and sequence homology to ESAT-6 and CFP-10, which induced dominant CD4+ T-cell responses in human tuberculosis (TB) models. We characterized T-cell responses including cytokine profiling, kinetics of activation, expansion, differentiation, TCR usage, and signaling of activation induced by Rv3615c compared with other M. tuberculosis-specific antigens. The expanded CD4+ T cells induced by Rv3615c predominately produced Th1, but less Th2 and Th17, cytokines and displayed effector/memory phenotypes (CD45RO+CD27-CD127-CCR7-). The magnitude of expansion and cytokine production was comparable to those induced by well-characterized the 6 kDa early secreted antigenic target (ESAT-6), the 10 kDa culture filtrate protein (CFP-10) and BCG. Rv3615c contained multiple epitopes Rv3615c1-15, Rv3615c6-20, Rv3615c66-80, Rv3615c71-85 and Rv3615c76-90 that activated CD4+ T cells. The Rv3615c-specific CD4+ T cells shared biased of T-cell receptor variable region of β chain (TCR Vβ) 1, 2, 4, 5.1, 7.1, 7.2 and/or 22 chains to promote their differentiation and proliferation respectively, by triggering a signaling cascade. Our data suggest that Rv3615c is a major target of Th1-type responses and can be a highly immunodominant antigen specific for M. tuberculosis infection.
Collapse
|
147
|
Pereira RM, Hogan PG, Rao A, Martinez GJ. Transcriptional and epigenetic regulation of T cell hyporesponsiveness. J Leukoc Biol 2017; 102:601-615. [PMID: 28606939 DOI: 10.1189/jlb.2ri0317-097r] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/08/2017] [Accepted: 05/10/2017] [Indexed: 12/19/2022] Open
Abstract
Naive CD8+ T cells differentiate into effector and memory cytolytic T cells (CTLs) during an acute infection. In contrast, in scenarios of persistent antigen stimulation, such as chronic infections and cancer, antigen-specific CTLs show a gradual decrease in effector function, a phenomenon that has been termed CD8+ T cell "exhaustion" or "dysfunction." Another hyporesponsive state, termed "anergy", is observed when T cells are activated in the absence of positive costimulatory signals. Among the many negative regulators induced in hyporesponsive T cells are inhibitory cell-surface receptors, such as PD-1, LAG-3, CTLA-4, and TIM-3; "checkpoint blockade" therapies that involve treatment of patients with cancer with blocking antibodies to those receptors show considerable promise in the clinic because the blocking antibodies can mitigate hyporesponsiveness and promote tumor rejection. In this review, we describe recent advances in our molecular understanding of these hyporesponsive states. We review evidence for the involvement of diverse transcription factors, metabolic programs, and chromatin accessibility changes in hyporesponsive T cells, and we discuss how checkpoint blockade therapies affect the molecular program of CD8+ T cell exhaustion.
Collapse
Affiliation(s)
- Renata M Pereira
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil;
| | - Patrick G Hogan
- Division of Signaling and Gene Expression, La Jolla Institute, San Diego, California, USA
| | - Anjana Rao
- Division of Signaling and Gene Expression, La Jolla Institute, San Diego, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA.,Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California, USA; and
| | - Gustavo J Martinez
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, Chicago Medical School, North Chicago, Illinois, USA
| |
Collapse
|
148
|
Aberrant plasma IL-7 and soluble IL-7 receptor levels indicate impaired T-cell response to IL-7 in human tuberculosis. PLoS Pathog 2017; 13:e1006425. [PMID: 28582466 PMCID: PMC5472333 DOI: 10.1371/journal.ppat.1006425] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 06/15/2017] [Accepted: 05/22/2017] [Indexed: 12/22/2022] Open
Abstract
T-cell proliferation and generation of protective memory during chronic infections depend on Interleukin-7 (IL-7) availability and receptivity. Regulation of IL-7 receptor (IL-7R) expression and signalling are key for IL-7-modulated T-cell functions. Aberrant expression of soluble (s) and membrane-associated (m) IL-7R molecules is associated with development of autoimmunity and immune failure in acquired immune deficiency syndrome (AIDS) patients. Here we investigated the role of IL-7/IL-7R on T-cell immunity in human tuberculosis. We performed two independent case-control studies comparing tuberculosis patients and healthy contacts. This was combined with follow-up examinations for a subgroup of tuberculosis patients under therapy and recovery. Blood plasma and T cells were characterised for IL-7/sIL-7R and mIL-7R expression, respectively. IL-7-dependent T-cell functions were determined by analysing STAT5 phosphorylation, antigen-specific cytokine release and by analysing markers of T-cell exhaustion and inflammation. Tuberculosis patients had lower soluble IL-7R (p < 0.001) and higher IL-7 (p < 0.001) plasma concentrations as compared to healthy contacts. Both markers were largely independent and aberrant expression normalised during therapy and recovery. Furthermore, tuberculosis patients had lower levels of mIL-7R in T cells caused by post-transcriptional mechanisms. Functional in vitro tests indicated diminished IL-7-induced STAT5 phosphorylation and impaired IL-7-promoted cytokine release of Mycobacterium tuberculosis-specific CD4+ T cells from tuberculosis patients. Finally, we determined T-cell exhaustion markers PD-1 and SOCS3 and detected increased SOCS3 expression during therapy. Only moderate correlation of PD-1 and SOCS3 with IL-7 expression was observed. We conclude that diminished soluble IL-7R and increased IL-7 plasma concentrations, as well as decreased membrane-associated IL-7R expression in T cells, reflect impaired T-cell sensitivity to IL-7 in tuberculosis patients. These findings show similarities to pathognomonic features of impaired T-cell functions and immune failure described in AIDS patients. IL-7 is important for the development and homeostasis of T cells and promotes antigen-specific T-cell responses. Aberrant expression of plasma IL-7 and soluble IL-7R are found in autoimmune diseases and chronic viral infections. In AIDS patients—especially those who fail to reconstitute T-cell numbers during therapy—impaired IL-7-promoted T-cell functions indicated T-cell exhaustion/senescence. In order to evaluate the potential impact of IL-7 on tuberculosis, we characterised various parameters involved in the IL-7-response of tuberculosis patients and healthy contacts. Despite IL-7 being available at higher plasma levels among tuberculosis patients, the T-cell response to IL-7 was impaired when compared to healthy contacts. Soluble IL-7R levels were aberrantly low in plasma during acute tuberculosis but did not account for impaired IL-7 usage. Chronic inflammation in tuberculosis patients—reflected by increased IL-6 plasma levels—did not account for dysfunctional T-cell responses and analysed T-cell exhaustion markers were only moderately correlated. Our findings demonstrate that availability of IL-7 alone is not sufficient to promote protective T-cell immunity against tuberculosis. We describe aberrant IL-7/soluble IL-7R expression and impaired IL-7-mediated T-cell functions in tuberculosis patients with similarities and differences to described IL-7 dysregulation seen in patients with AIDS.
Collapse
|
149
|
Gao Q, Dong X, Luo Y, Zhang G, Shan J, Wang Q, He Q, Zhang L, Wang J, Zhu B, Ma X. Construction of human MASP-2-CCP1/2SP, CCP2SP, SP plasmid DNA nanolipoplexes and the effects on tuberculosis in BCG-infected mice. Microb Pathog 2017; 109:200-208. [PMID: 28578092 DOI: 10.1016/j.micpath.2017.05.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 12/25/2022]
Abstract
The lectin pathway, one of the complement cascade systems, provides the primary line of defense against invading pathogens. The serine protease of MASP-2 plays an essential role in complement activation of the lectin pathway. The C-terminal segment of MASP-2 is comprised of the CCP1-CCP2-SP domains, and is the crucial catalytic segment. However, what is the effect of CCP1-CCP2-SP domains in controlling chronic infection is unknown. In order to evaluate the potential impact of CCP1-CCP2-SP domains on tuberculosis, we constructed the human MASP-2 CCP1/2SP, CCP2SP and SP recombinant plasmids, and delivered these plasmids by DNA-DOTAP:cholesterol cationic nanolipoplexes to BCG-infected mice. After 21 days post DNA-DOTAP:chol nanolipoplexes application, we analyzed bacteria loads of pulmonary, pathology of granuloma, lymphocyte subpopulations. The C3a, C4a and MASP-2 levels in serum were measured with enzyme-linked immunosorbent assays. Compared to the control group that received GFP DNA-DOTAP:chol nanolipoplexes, MASP-2 CCP1/2SP DNA-DOTAP:chol nanolipoplexes treated group showed significantly enlarged pulmonary granulomas lesion (P < 0.05) and did not reduce bacteria loads in the lung tissue (P < 0.05). Furthermore, the levels of C3a in serum were decreased (P < 0.05), the number and percentage of PD1+ and Tim3+ cells subgroups were increased in BCG-infected mice after treated with MASP-2 CCP1/2SP DNA-DOTAP:chol nanolipoplexes (P < 0.05). But, there was no statistical difference in the serum C4a and MASP-2 level among DNA nanolipoplexes treated groups (P > 0.05). These findings provided experimental evidence that MASP-2 CCP1/2SP DNA nanolipoplexes shown the negative efficacy in controlling Mycobacterium tuberculosis infection, and displayed a potential role of down-regulating T-cell-mediated immunity in tuberculosis.
Collapse
Affiliation(s)
- Qi Gao
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Xinfang Dong
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yanping Luo
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Guochao Zhang
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jinyu Shan
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester LE1 9HN, UK
| | - Qian Wang
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Qi He
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Lifeng Zhang
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jingqiu Wang
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Bingdong Zhu
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Xingming Ma
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Key Lab of Preclinical Study for New Drugs of Gansu Province, Lanzhou, 730000, China.
| |
Collapse
|
150
|
Innate and Adaptive Immune Defects in Chronic Pulmonary Aspergillosis. J Fungi (Basel) 2017; 3:jof3020026. [PMID: 29371544 PMCID: PMC5715918 DOI: 10.3390/jof3020026] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 12/17/2022] Open
Abstract
We evaluated the expression of biomarkers of innate and adaptive immune response in correlation with underlying conditions in 144 patients with chronic pulmonary aspergillosis (CPA). Patients with complete medical and radiological records, white cell counts, and a complete panel of CD3, CD4, CD8, CD19, and CD56 lymphocyte subsets were included. Eighty-four (58%) patients had lymphopenia. Six (4%) patients had lymphopenia in all five CD variables. There were 62 (43%) patients with low CD56 and 62 (43%) patients with low CD19. Ten (7%) patients had isolated CD19 lymphopenia, 18 (13%) had isolated CD56 lymphopenia, and 15 (10%) had combined CD19 and CD56 lymphopenia only. Forty-eight (33%) patients had low CD3 and 46 (32%) had low CD8 counts. Twenty-five (17%) patients had low CD4, 15 (10%) of whom had absolute CD4 counts <200/μL. Multivariable logistic regression showed associations between: low CD19 and pulmonary sarcoidosis (Odds Ratio (OR), 5.53; 95% Confidence Interval (CI), 1.43–21.33; p = 0.013), and emphysema (OR, 4.58; 95% CI; 1.36–15.38; p = 0.014), low CD56 and no bronchiectasis (OR, 0.27; 95% CI, 0.10–0.77; p = 0.014), low CD3 and both multicavitary CPA disease (OR, 2.95; 95% CI, 1.30–6.72; p = 0.010) and pulmonary sarcoidosis (OR, 4.94; 95% CI, 1.39–17.57; p = 0.014). Several subtle immune defects are found in CPA.
Collapse
|