1
|
Romero-Cruz VA, Ramos-Ligonio A, García-Alejandro K, Cerecedo-García M, Lagunes-Castro MDLS, López-Monteon A. Immunization of recombinant NS3 protein (protease region) of dengue virus induces high levels of CTLA-4 and apoptosis in splenocytes of BALB/c mice. Virus Genes 2024; 60:475-487. [PMID: 39102085 DOI: 10.1007/s11262-024-02095-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024]
Abstract
DENV infection outcomes depend on the host's variable expression of immune receptors and mediators, leading to either resolution or exacerbation. While the NS3 protein is known to induce robust immune responses, the specific impact of its protease region epitopes remains unclear. This study investigated the effect of recombinant NS3 protease region proteins from all four DENV serotypes on splenocyte activation in BALB/c mice (n = 5/group). Mice were immunized with each protein, and their splenocytes were subsequently stimulated with homologous antigens. We measured the expression of costimulatory molecules (CD28, CD80, CD86, CD152) by flow cytometry, along with IL-2 production, CD25 expression, and examined the antigen-specific activation of CD4 + and CD8 + T cells. Additionally, the expression of IL-1, IL-10, and TGF-β1 in splenocytes from immunized animals was assessed. Apoptosis was evaluated using Annexin V/PI staining and DNA fragmentation analysis. Stimulation of splenocytes from immunized mice triggered apoptosis (phosphatidylserine exposure and caspase 3/7 activation) and increased costimulatory molecule expression, particularly CD152. Low IL-2 production and low CD25 expression, as well as sustained expression of the IL-10 gene. These results suggest that these molecules might be involved in mechanisms by which the NS3 protein contributes to viral persistence and disease pathogenesis.
Collapse
Affiliation(s)
- Víctor Adolfo Romero-Cruz
- Doctorado en Ciencias Biomédicas, Universidad Veracruzana, Luis Castelazo, Animas, 91190, Xalapa, Veracruz, Mexico
| | - Angel Ramos-Ligonio
- LADISER Inmunología y Biología Molecular, Facultad de Ciencias Químicas, Edificio D, Universidad Veracruzana, Prolongación de Oriente 6 No. 1009, Col. Rafael Alvarado, 94340, Orizaba, Veracruz, Mexico
| | - Karen García-Alejandro
- LADISER Inmunología y Biología Molecular, Facultad de Ciencias Químicas, Edificio D, Universidad Veracruzana, Prolongación de Oriente 6 No. 1009, Col. Rafael Alvarado, 94340, Orizaba, Veracruz, Mexico
- Maestría en Procesos Biológicos, Universidad Veracruzana, Prolongación de Oriente 6 No. 1009, Col. Rafael Alvarado, 94340, Orizaba, Veracruz, Mexico
| | - Melissa Cerecedo-García
- LADISER Inmunología y Biología Molecular, Facultad de Ciencias Químicas, Edificio D, Universidad Veracruzana, Prolongación de Oriente 6 No. 1009, Col. Rafael Alvarado, 94340, Orizaba, Veracruz, Mexico
- Maestría en Procesos Biológicos, Universidad Veracruzana, Prolongación de Oriente 6 No. 1009, Col. Rafael Alvarado, 94340, Orizaba, Veracruz, Mexico
| | - María de la Soledad Lagunes-Castro
- LADISER Inmunología y Biología Molecular, Facultad de Ciencias Químicas, Edificio D, Universidad Veracruzana, Prolongación de Oriente 6 No. 1009, Col. Rafael Alvarado, 94340, Orizaba, Veracruz, Mexico
| | - Aracely López-Monteon
- LADISER Inmunología y Biología Molecular, Facultad de Ciencias Químicas, Edificio D, Universidad Veracruzana, Prolongación de Oriente 6 No. 1009, Col. Rafael Alvarado, 94340, Orizaba, Veracruz, Mexico.
| |
Collapse
|
2
|
Nikoo M, Hassan ZF, Mardasi M, Rostamnezhad E, Roozbahani F, Rahimi S, Mohammadi J. Hepatocellular carcinoma (HCC) immunotherapy by anti-PD-1 monoclonal antibodies: A rapidly evolving strategy. Pathol Res Pract 2023; 247:154473. [PMID: 37207558 DOI: 10.1016/j.prp.2023.154473] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest cancers in the world, with a high relapse rate. Delayed symptom onset observed in 70-80% of patients leads to diagnosis in advanced stages commonly associated with chronic liver disease. Programmed cell death protein 1 (PD-1) blockade therapy has recently emerged as a promising therapeutic option in the clinical management of several advanced malignancies, including HCC, due to the activation of exhausted tumor-infiltrating lymphocytes and improved outcomes of T-cell function. However, many people with HCC do not respond to PD-1 blockade therapy, and the diversity of immune-related adverse events (irAEs) restricts their clinical utility. Therefore, numerous effective combinatory strategies, including combinations with anti-PD-1 antibodies and other therapeutic methods ranging from chemotherapy to targeted therapies, are evolving to improve therapeutic outcomes and evoke synergistic anti-tumor impressions in patients with advanced HCC. Unfortunately, combined therapy may have more side effects than single-agent treatment. Nonetheless, identifying appropriate predictive biomarkers can aid in managing potential immune-related adverse events by distinguishing patients who respond best to PD-1 inhibitors as single agents or in combination strategies. In the present review, we summarize the therapeutic potential of PD-1 blockade therapy for advanced HCC patients. Besides, a glimpse of the pivotal predictive biomarkers influencing a patient's response to anti-PD-1 antibodies will be provided.
Collapse
Affiliation(s)
- Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Mahsa Mardasi
- Biotechnology Department, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University G. C., Evin, Tehran, Iran
| | - Elmira Rostamnezhad
- Department of Molecular Genetics, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Fatemeh Roozbahani
- Department of Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sahel Rahimi
- Industrial and Environmental Biotechnology Department, National Institute of Genetic Engineering and Biotechnology(NIGEB), Tehran, Iran
| | - Javad Mohammadi
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran.
| |
Collapse
|
3
|
Elsegood CL, Tirnitz-Parker JE, Olynyk JK, Yeoh GC. Immune checkpoint inhibition: prospects for prevention and therapy of hepatocellular carcinoma. Clin Transl Immunology 2017; 6:e161. [PMID: 29326816 PMCID: PMC5704099 DOI: 10.1038/cti.2017.47] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 08/10/2017] [Accepted: 09/10/2017] [Indexed: 02/06/2023] Open
Abstract
The global prevalence of liver cancer is rapidly rising, mostly as a result of the amplified incidence rates of viral hepatitis, alcohol abuse and obesity in recent decades. Treatment options for liver cancer are remarkably limited with sorafenib being the gold standard for advanced, unresectable hepatocellular carcinoma but offering extremely limited improvement of survival time. The immune system is now recognised as a key regulator of cancer development through its ability to protect against infection and chronic inflammation, which promote cancer development, and eliminate tumour cells when present. However, the tolerogenic nature of the liver means that the immune response to infection, chronic inflammation and tumour cells within the hepatic environment is usually ineffective. Here we review the roles that immune cells and cytokines have in the development of the most common primary liver cancer, hepatocellular carcinoma (HCC). We then examine how the immune system may be subverted throughout the stages of HCC development, particularly with respect to immune inhibitory molecules, also known as immune checkpoints, such as programmed cell death protein-1, programmed cell death 1 ligand 1 and cytotoxic T lymphocyte antigen 4, which have become therapeutic targets. Finally, we assess preclinical and clinical studies where immune checkpoint inhibitors have been used to modify disease during the carcinogenic process. In conclusion, inhibitory molecule-based immunotherapy for HCC is in its infancy and further detailed research in relevant in vivo models is required before its full potential can be realised.
Collapse
Affiliation(s)
- Caryn L Elsegood
- School of Biomedical Science, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Janina Ee Tirnitz-Parker
- School of Biomedical Science, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - John K Olynyk
- School of Biomedical Science, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,Department of Gastroenterology and Hepatology, Fiona Stanley and Fremantle Hospitals, South Metropolitan Health Service, Murdoch, Western Australia, Australia.,School of Health and Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - George Ct Yeoh
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia, Australia.,Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
4
|
Schaenman J, Korin Y, Sidwell T, Kandarian F, Harre N, Gjertson D, Lum E, Reddy U, Huang E, Pham P, Bunnapradist S, Danovitch G, Veale J, Gritsch H, Reed E. Increased Frequency of BK Virus-Specific Polyfunctional CD8+ T Cells Predict Successful Control of BK Viremia After Kidney Transplantation. Transplantation 2017; 101:1479-1487. [PMID: 27391197 PMCID: PMC5219876 DOI: 10.1097/tp.0000000000001314] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND BK virus infection remains an important cause of loss of allograft function after kidney transplantation. We sought to determine whether polyfunctional T cells secreting multiple cytokines simultaneously, which have been shown to be associated with viral control, could be detected early after start of BK viremia, which would provide insight into the mechanism of successful antiviral control. METHODS Peripheral blood mononuclear cells collected during episodes of BK viral replication were evaluated by multiparameter flow cytometry after stimulation by overlapping peptide pools of BK virus antigen to determine frequency of CD8+ and CD4+ T cells expressing 1 or more cytokines simultaneously, as well as markers of T-cell activation, exhaustion, and maturation. RESULTS BK virus controllers, defined as those with episodes of BK viremia of 3 months or less, had an 11-fold increase in frequency of CD8+ polyfunctional T cells expressing multiple cytokines, as compared with patients with prolonged episodes of BK viremia. Patients with only low level BK viremia expressed low frequencies of polyfunctional T cells. Polyfunctional T cells were predominantly of the effector memory maturation subtype and expressed the cytotoxicity marker CD107a. CONCLUSIONS Noninvasive techniques for immune assessment of peripheral blood can provide insight into the mechanism of control of BK virus replication and may allow for future patient risk stratification and customization of immune suppression at the onset of BK viremia.
Collapse
Affiliation(s)
- J.M. Schaenman
- Division of Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Y. Korin
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - T. Sidwell
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - F. Kandarian
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - N. Harre
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - D. Gjertson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Department of Biostatistics, UCLA School of Public Health, Los Angeles, CA
| | - E. Lum
- Division of Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - U. Reddy
- Division of Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - E. Huang
- Division of Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - P.T. Pham
- Division of Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - S. Bunnapradist
- Division of Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - G. Danovitch
- Division of Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - J. Veale
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - H.A. Gritsch
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - E.F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
5
|
Klein M, Schmalzing M, Almanzar G, Benoit S, Hamm H, Tony HP, Goebeler M, Prelog M. Contribution of CD8+ T cells to inflammatory cytokine production in systemic sclerosis (SSc). Autoimmunity 2016; 49:532-546. [PMID: 27560622 DOI: 10.1080/08916934.2016.1217997] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Only limited attention has been paid to the role of CD8 + T cells in the etiopathogenesis and progression of systemic sclerosis (SSc). CD8 + T cells may have autoantigen-specific and pro-inflammatory but also immunomodulatory properties. To investigate the differentiation of CD8 + T cells, staining of cell surface factors and of chemokine receptors were performed. In addition, the cytokine-producing ability of circulating CD8 + T cells and their sensitivity to suppression by regulatory T cells (Tregs) were compared between patients with diffuse (dcSSc) or limited cutaneous SSc (lcSSc) and healthy individuals. We identified CD8 + T cells as producers of pro-inflammatory type-2 cytokines with a significant contribution of memory CD8 + T cells. Memory CD8 + T cells of SSc patients stayed unaltered after suppression with autologous Tregs. Expression of chemokine receptors was significantly correlated with intracellular cytokine production in CD8 + T cells with a clear dichotomy of type 1 and type 2 cytokines. High levels of intracellular cytokines, such as interleukin-(IL)-4, IL-13 and tumor-necrosis-factor-alpha (TNFalpha) were positively associated with the presence of Scl-70 or anti-centromere antibodies and negatively with the administration of glucocorticoids. Administration of glucocorticoids was positively associated with higher IFNgamma production. Lack of anti-centromere antibodies and therapy with methotrexate were positively associated with higher intracellular IL-10 production. CD8 + T cells may significantly contribute to inflammation in SSc. Our findings suggest to not only focus on T helper cells in the development of therapeutic strategies but also to consider the role of CD8 + T cells in the etiopathogenesis and perpetuation of SSc.
Collapse
Affiliation(s)
- Matthias Klein
- a Department of Pediatrics, Pediatric Rheumatology and Special Immunology , University Hospital Wuerzburg , Wuerzburg , Germany
| | - Marc Schmalzing
- b Department of Internal Medicine II, Division of Rheumatology and Clinical Immunology , University Hospital Wuerzburg , Wuerzburg , Germany , and
| | - Giovanni Almanzar
- a Department of Pediatrics, Pediatric Rheumatology and Special Immunology , University Hospital Wuerzburg , Wuerzburg , Germany
| | - Sandrine Benoit
- c Department of Dermatology, Venereology and Allergology , University Hospital Wuerzburg , Wuerzburg , Germany
| | - Henning Hamm
- c Department of Dermatology, Venereology and Allergology , University Hospital Wuerzburg , Wuerzburg , Germany
| | - Hans-Peter Tony
- b Department of Internal Medicine II, Division of Rheumatology and Clinical Immunology , University Hospital Wuerzburg , Wuerzburg , Germany , and
| | - Matthias Goebeler
- c Department of Dermatology, Venereology and Allergology , University Hospital Wuerzburg , Wuerzburg , Germany
| | - Martina Prelog
- a Department of Pediatrics, Pediatric Rheumatology and Special Immunology , University Hospital Wuerzburg , Wuerzburg , Germany
| |
Collapse
|
6
|
Cunningham CR, Champhekar A, Tullius MV, Dillon BJ, Zhen A, de la Fuente JR, Herskovitz J, Elsaesser H, Snell LM, Wilson EB, de la Torre JC, Kitchen SG, Horwitz MA, Bensinger SJ, Smale ST, Brooks DG. Type I and Type II Interferon Coordinately Regulate Suppressive Dendritic Cell Fate and Function during Viral Persistence. PLoS Pathog 2016; 12:e1005356. [PMID: 26808628 PMCID: PMC4726812 DOI: 10.1371/journal.ppat.1005356] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/01/2015] [Indexed: 12/21/2022] Open
Abstract
Persistent viral infections are simultaneously associated with chronic inflammation and highly potent immunosuppressive programs mediated by IL-10 and PDL1 that attenuate antiviral T cell responses. Inhibiting these suppressive signals enhances T cell function to control persistent infection; yet, the underlying signals and mechanisms that program immunosuppressive cell fates and functions are not well understood. Herein, we use lymphocytic choriomeningitis virus infection (LCMV) to demonstrate that the induction and functional programming of immunosuppressive dendritic cells (DCs) during viral persistence are separable mechanisms programmed by factors primarily considered pro-inflammatory. IFNγ first induces the de novo development of naive monocytes into DCs with immunosuppressive potential. Type I interferon (IFN-I) then directly targets these newly generated DCs to program their potent T cell immunosuppressive functions while simultaneously inhibiting conventional DCs with T cell stimulating capacity. These mechanisms of monocyte conversion are constant throughout persistent infection, establishing a system to continuously interpret and shape the immunologic environment. MyD88 signaling was required for the differentiation of suppressive DCs, whereas inhibition of stimulatory DCs was dependent on MAVS signaling, demonstrating a bifurcation in the pathogen recognition pathways that promote distinct elements of IFN-I mediated immunosuppression. Further, a similar suppressive DC origin and differentiation was also observed in Mycobacterium tuberculosis infection, HIV infection and cancer. Ultimately, targeting the underlying mechanisms that induce immunosuppression could simultaneously prevent multiple suppressive signals to further restore T cell function and control persistent infections. Persistent virus infections induce host derived immunosuppressive factors that attenuate the immune response and prevent control of infection. Although the mechanisms of T cell exhaustion are being defined, we know surprisingly little about the underlying mechanisms that induce the immunosuppressive state and the origin and functional programming of the cells that deliver these signals to the T cells. We recently demonstrated that type I interferon (IFN-I) signaling was responsible for many of the immune dysfunctions associated with persistent virus infection and in particular the induced expression of the suppressive factors IL-10 and PDL1 by dendritic cells (DCs). Yet, mechanistically how IFN-I signaling specifically generates and programs cells to become immunosuppressive is still unknown. Herein, we define the underlying mechanisms of IFN-I mediated immunosuppression and establish that the induction of factors and the generation of the DCs that express them are separable events integrally reliant on additional inflammatory factors. Further, we demonstrate a similar derivation of the suppressive DCs that emerge in other diseases associated with prolonged inflammation and immunosuppression, specifically in HIV infection, Mycobacterium tuberculosis, and cancer, indicating a conserved origin of immunosuppression and suggesting that targeting the pathways that underlie expression of immunosuppressive cells and factors could be beneficial to treat multiple chronic diseases.
Collapse
Affiliation(s)
- Cameron R. Cunningham
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Ameya Champhekar
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Michael V. Tullius
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Barbara Jane Dillon
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Anjie Zhen
- Division of Hematology and Oncology, Department of Medicine, UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Justin Rafael de la Fuente
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Jonathan Herskovitz
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Heidi Elsaesser
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Princess Margaret Cancer Center, Immune Therapy Program, University Health Network, Toronto, Ontario
| | - Laura M. Snell
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Princess Margaret Cancer Center, Immune Therapy Program, University Health Network, Toronto, Ontario
| | - Elizabeth B. Wilson
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Juan Carlos de la Torre
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Scott G. Kitchen
- Division of Hematology and Oncology, Department of Medicine, UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Marcus A. Horwitz
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Steven J. Bensinger
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Stephen T. Smale
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - David G. Brooks
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Princess Margaret Cancer Center, Immune Therapy Program, University Health Network, Toronto, Ontario
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
7
|
Puntambekar SS, Hinton DR, Yin X, Savarin C, Bergmann CC, Trapp BD, Stohlman SA. Interleukin-10 is a critical regulator of white matter lesion containment following viral induced demyelination. Glia 2015; 63:2106-2120. [PMID: 26132901 PMCID: PMC4755156 DOI: 10.1002/glia.22880] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 06/08/2015] [Accepted: 06/08/2015] [Indexed: 12/29/2022]
Abstract
Neurotropic coronavirus induces an acute encephalomyelitis accompanied by focal areas of demyelination distributed randomly along the spinal column. The initial areas of demyelination increase only slightly after the control of infection. These circumscribed focal lesions are characterized by axonal sparing, myelin ingestion by macrophage/microglia, and glial scars associated with hypertrophic astrocytes, which proliferate at the lesion border. Accelerated virus control in mice lacking the anti‐inflammatory cytokine IL‐10 was associated with limited initial demyelination, but low viral mRNA persistence similar to WT mice and declining antiviral cellular immunity. Nevertheless, lesions exhibited sustained expansion providing a model of dysregulated white matter injury temporally remote from the acute CNS insult. Expanding lesions in the absence of IL‐10 are characterized by sustained microglial activation and partial loss of macrophage/microglia exhibiting an acquired deactivation phenotype. Furthermore, IL‐10 deficiency impaired astrocyte organization into mesh like structures at the lesion borders, but did not prevent astrocyte hypertrophy. The formation of discrete foci of demyelination in IL‐10 sufficient mice correlated with IL‐10 receptor expression exclusively on astrocytes in areas of demyelination suggesting a critical role for IL‐10 signaling to astrocytes in limiting expansion of initial areas of white matter damage. GLIA 2015;63:2106–2120
Collapse
Affiliation(s)
- Shweta S Puntambekar
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
| | - David R Hinton
- Department of Pathology, The University of Southern California Keck School of Medicine, Los Angeles, California
| | - Xinghua Yin
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Carine Savarin
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Cornelia C Bergmann
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Stephen A Stohlman
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
8
|
Yamada DH, Elsaesser H, Lux A, Timmerman JM, Morrison SL, de la Torre JC, Nimmerjahn F, Brooks DG. Suppression of Fcγ-receptor-mediated antibody effector function during persistent viral infection. Immunity 2015; 42:379-390. [PMID: 25680277 DOI: 10.1016/j.immuni.2015.01.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/21/2014] [Accepted: 12/31/2014] [Indexed: 01/06/2023]
Abstract
Understanding how viruses subvert host immunity and persist is essential for developing strategies to eliminate infection. T cell exhaustion during chronic viral infection is well described, but effects on antibody-mediated effector activity are unclear. Herein, we show that increased amounts of immune complexes generated in mice persistently infected with lymphocytic choriomeningitis virus (LCMV) suppressed multiple Fcγ-receptor (FcγR) functions. The high amounts of immune complexes suppressed antibody-mediated cell depletion, therapeutic antibody-killing of LCMV infected cells and human CD20-expressing tumors, as well as reduced immune complex-mediated cross-presentation to T cells. Suppression of FcγR activity was not due to inhibitory FcγRs or high concentrations of free antibody, and proper FcγR functions were restored when persistently infected mice specifically lacked immune complexes. Thus, we identify a mechanism of immunosuppression during viral persistence with implications for understanding effective antibody activity aimed at pathogen control.
Collapse
Affiliation(s)
- Douglas H Yamada
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Heidi Elsaesser
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Anja Lux
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erwin-Rommelstrasse 3, 91058, Erlangen, Germany
| | - John M Timmerman
- Division of Hematology & Oncology, Department of Medicine, and Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA 90095, USA
| | - Sherie L Morrison
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Juan Carlos de la Torre
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Falk Nimmerjahn
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erwin-Rommelstrasse 3, 91058, Erlangen, Germany
| | - David G Brooks
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA; UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
9
|
Reyes-Cerpa S, Reyes-López F, Toro-Ascuy D, Montero R, Maisey K, Acuña-Castillo C, Sunyer JO, Parra D, Sandino AM, Imarai M. Induction of anti-inflammatory cytokine expression by IPNV in persistent infection. FISH & SHELLFISH IMMUNOLOGY 2014; 41:172-182. [PMID: 25193394 DOI: 10.1016/j.fsi.2014.08.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 08/18/2014] [Accepted: 08/25/2014] [Indexed: 06/03/2023]
Abstract
Infectious Pancreatic Necrosis Virus (IPNV) is the agent of a well-characterized acute disease that produces a systemic infection and high mortality in farmed fish species but also persistent infection in surviving fish after outbreaks. Because viral persistence of susceptible mammal hosts appears to be associated with the modulation of anti-inflammatory cytokine expression, in this study we examined the expression levels of key pro- and anti-inflammatory cytokines in kidney and spleen of trout, as well as humoral immune response (IgM and IgT) during experimental persistent viral infection and in the acute phase of infection as a comparison. IPNV infection in rainbow trout resulted in a distinct profile of cytokine expression depending on the type of infection, acute or persistent. Levels of early pro-inflammatory cytokines, IL-1β and IL-8, did not increase in the head kidney of the fish with persistent asymptomatic infection but increased in some of the symptomatic infected fish. The antiviral cytokine IFNα was not significantly induced in any of the infected fish groups. The level of expression of the Th1-related cytokine IL-12 was significantly higher in trout with persistent asymptomatic infection than in symptomatic fish. This was also accompanied by an increase in IFNγ. The anti-inflammatory cytokines IL-10 and TGF-β1 had distinct expression profiles. While IL-10 expression increased in all infected fish, TGF-β1 was only up-regulated in fish with persistent infection. All infected fish had significantly lower total IgM levels than the non-infected fish whereas IgT levels did not change. Specific and neutralizing antibodies against IPNV were not observed in acute and persistent infection except in the group of fish with the lowest degree of clinical signs. Interestingly, the lack of humoral immune response could be associated with the high expression of anti-inflammatory cytokines, which might inhibit antibody production. The balance between pro-inflammatory Th1 type cytokines and the regulatory cytokines could explain the high percentage of survival and the resolution of the inflammatory response in the IPNV-infected fish but also the establishment of viral persistence.
Collapse
Affiliation(s)
- Sebastián Reyes-Cerpa
- Laboratorio de Inmunología, Centro de Biotecnología Acuícola (CBA), Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.
| | - Felipe Reyes-López
- Laboratorio de Inmunología, Centro de Biotecnología Acuícola (CBA), Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Daniela Toro-Ascuy
- Laboratorio de Inmunología, Centro de Biotecnología Acuícola (CBA), Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Ruth Montero
- Laboratorio de Inmunología, Centro de Biotecnología Acuícola (CBA), Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Kevin Maisey
- Laboratorio de Inmunología, Centro de Biotecnología Acuícola (CBA), Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Claudio Acuña-Castillo
- Laboratorio de Inmunoterapia, Centro de Biotecnología Acuícola (CBA), Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - J Oriol Sunyer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Parra
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ana María Sandino
- Laboratorio de Virología, Centro de Biotecnología Acuícola (CBA), Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Mónica Imarai
- Laboratorio de Inmunología, Centro de Biotecnología Acuícola (CBA), Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|
10
|
Type I interferon suppresses de novo virus-specific CD4 Th1 immunity during an established persistent viral infection. Proc Natl Acad Sci U S A 2014; 111:7409-14. [PMID: 24799699 DOI: 10.1073/pnas.1401662111] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
CD4 T cells are central to orchestrate, sustain, and potentially regenerate antiviral immunity throughout persistent viral infections. Although the evolving immune environment during persistent infection reshapes established CD4 T-cell responses, the fate of naïve CD4 T cells primed in the midst of persistent infection is unclear. We demonstrate that, in marked contrast to the onset of infection, virus-specific CD4 T cells primed during an established persistent infection have diminished ability to develop Th1 responses, to efficiently accumulate in peripheral tissues, and almost exclusively differentiate into T follicular helper cells. Consistent with suppressed Th1 and heightened Tfh differentiation, virus-specific CD4 T cells primed during the established persistent infection provide help to B cells, but only limited help to CD8 T cells. The suppression of de novo Th1 generation and tissue distribution was mediated by chronic type I IFN (IFN-I) production and was effectively restored by blocking IFN-I signaling during CD4 T-cell priming. Thus, we establish a suppressive function of chronic IFN-I signaling and mechanism of immunoregulation during an established persistent virus infection.
Collapse
|
11
|
Abstract
Interleukin-10 (IL-10) is a key immunoregulatory cytokine that functions to prevent inflammatory and autoimmune diseases. Despite the critical role for IL-10 produced by effector CD8(+) T cells during pathogen infection and autoimmunity, the mechanisms regulating its production are poorly understood. We show that loss of the inhibitor of DNA binding 2 (Id2) in T cells resulted in aberrant IL-10 expression in vitro and in vivo during influenza virus infection and in a model of acute graft-versus-host disease (GVHD). Furthermore, IL-10 overproduction substantially reduced the immunopathology associated with GVHD. We demonstrate that Id2 acts to repress the E2A-mediated trans-activation of the Il10 locus. Collectively, our findings uncover a key regulatory role of Id2 during effector T cell differentiation necessary to limit IL-10 production by activated T cells and minimize their suppressive activity during the effector phase of disease control.
Collapse
|
12
|
Ng CT, Oldstone MBA. IL-10: achieving balance during persistent viral infection. Curr Top Microbiol Immunol 2014; 380:129-44. [PMID: 25004816 DOI: 10.1007/978-3-662-43492-5_6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The clearance of viral infections is reliant on the coordination and balance of inflammatory factors necessary for viral destruction and immunoregulatory mechanisms necessary to prevent host pathology. In the case of persistent viral infections, immunoregulatory pathways prevent the immune response from clearing the virus, resulting in a long-term equilibrium between host and pathogen. Consequently, negative immune regulators are being considered as a therapeutic target to treat persistent and chronic viral infections. In this review, we will highlight the current understanding of the important negative immune regulator interleukin-10 (IL-10) in persistent viral infection. Though its main role for the host is to limit immune-mediated pathology, IL-10 is a multifunctional cytokine that differentially regulates a number of different hematopoietic cell types. IL-10 has been shown to play a role in a number of infectious diseases and many viral pathogens specifically exploit the IL-10 pathway to help evade host immunity. Recent advances have demonstrated that manipulation of IL-10 signaling during persistent viral infection can alter T cell responses in vivo and that this manipulation can lead to the clearance of persistent viral infection. Furthermore, there have been crucial advances in the understanding of factors that induce IL-10. We summarize lessons learned about IL-10 in model organisms and human persistent infections and conclude with the potential use of IL-10 to treat persistent viral infections.
Collapse
Affiliation(s)
- Cherie T Ng
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | | |
Collapse
|
13
|
Ng CT, Snell LM, Brooks DG, Oldstone MBA. Networking at the level of host immunity: immune cell interactions during persistent viral infections. Cell Host Microbe 2013; 13:652-64. [PMID: 23768490 DOI: 10.1016/j.chom.2013.05.014] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Persistent viral infections are the result of a series of connected events that culminate in diminished immunity and the inability to eliminate infection. By building our understanding of how distinct components of the immune system function both individually and collectively in productive versus abortive responses, new potential therapeutic targets can be developed to overcome immune dysfunction and thus fight persistent infections. Using lymphocytic choriomeningitis virus (LCMV) as a model of a persistent virus infection and drawing parallels to persistent human viral infections such as human immunodeficiency virus (HIV) and hepatitis C virus (HCV), we describe the cellular relationships and interactions that determine the outcome of initial infection and highlight immune targets for therapeutic intervention to prevent or treat persistent infections. Ultimately, these findings will further our understanding of the immunologic basis of persistent viral infection and likely lead to strategies to treat human viral infections.
Collapse
Affiliation(s)
- Cherie T Ng
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
14
|
Expression of inhibitory markers is increased on effector memory T cells during hepatitis C virus/HIV coinfection as compared to hepatitis C virus or HIV monoinfection. AIDS 2013; 27:2191-200. [PMID: 23820090 DOI: 10.1097/qad.0b013e32836285e4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Hepatitis C virus (HCV)/HIV coinfection is associated with rapid progression of hepatic fibrosis and liver disease. T-cell response has been implicated in the pathophysiological outcome of the disease. DESIGN This study sought to evaluate the role of memory T-cell exhaustion in enhancing immune dysfunction during coinfection. METHODS Sixty-four patients were included in the study; HCV monoinfected (n = 21), HIV monoinfected (n = 23), HCV/HIV coinfected (n = 20), and healthy controls (n = 20). Peripheral blood mononuclear cells (PBMCs) were isolated; immunophenotyped and functional assays were performed. RESULTS A significant increase in the naive T cells and central memory T cells and a marked reduction in effector memory T cells (TEM) were observed with coinfection as compared to monoinfection. Inhibitory markers programmed death 1 (PD-1) and T-cell immunoglobulin and mucin domain containing molecule 3 (TIM3) were highly upregulated on TEM in coinfection and functionally, these TEM cells displayed lowered proliferation. Increased expression of PD-1 and TIM3 correlated with decreased levels of CD8+CD107a+ TEM cells in coinfection. Pro-inflammatory cytokines interferon-γ and interleukin-2 (IL-2) secretion by TEM cells were also reduced during chronic viral infection. Secretion of IL-10, a human cytokine synthesis inhibitory factor, was significantly upregulated in CD4 TEM with HCV/HIV coinfection in comparison to HCV monoinfection. CONCLUSION TEM cells play an important role during viral infection and enhanced expression of inhibitory markers is associated with decreased proliferation and cytotoxicity and increased IL-10 production, which was pronounced in HCV/HIV coinfection. Thus, decreased TEM functionality contributes to diminished host immune responses during HCV/HIV coinfection as compared to HCV or HIV monoinfection.
Collapse
|
15
|
Riedel A, Mofolo B, Avota E, Schneider-Schaulies S, Meintjes A, Mulder N, Kneitz S. Accumulation of splice variants and transcripts in response to PI3K inhibition in T cells. PLoS One 2013; 8:e50695. [PMID: 23383294 PMCID: PMC3562341 DOI: 10.1371/journal.pone.0050695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 10/23/2012] [Indexed: 12/17/2022] Open
Abstract
Background Measles virus (MV) causes T cell suppression by interference with phosphatidylinositol-3-kinase (PI3K) activation. We previously found that this interference affected the activity of splice regulatory proteins and a T cell inhibitory protein isoform was produced from an alternatively spliced pre-mRNA. Hypothesis Differentially regulated and alternatively splice variant transcripts accumulating in response to PI3K abrogation in T cells potentially encode proteins involved in T cell silencing. Methods To test this hypothesis at the cellular level, we performed a Human Exon 1.0 ST Array on RNAs isolated from T cells stimulated only or stimulated after PI3K inhibition. We developed a simple algorithm based on a splicing index to detect genes that undergo alternative splicing (AS) or are differentially regulated (RG) upon T cell suppression. Results Applying our algorithm to the data, 9% of the genes were assigned as AS, while only 3% were attributed to RG. Though there are overlaps, AS and RG genes differed with regard to functional regulation, and were found to be enriched in different functional groups. AS genes targeted extracellular matrix (ECM)-receptor interaction and focal adhesion pathways, while RG genes were mainly enriched in cytokine-receptor interaction and Jak-STAT. When combined, AS/RG dependent alterations targeted pathways essential for T cell receptor signaling, cytoskeletal dynamics and cell cycle entry. Conclusions PI3K abrogation interferes with key T cell activation processes through both differential expression and alternative splicing, which together actively contribute to T cell suppression.
Collapse
Affiliation(s)
- Alice Riedel
- Institute for Virology and Immunobiology, University of Wuerzburg, Versbacher, Wuerzburg, Germany
| | - Boitumelo Mofolo
- Computational Biology Group, Department of Clinical Laboratory Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Faculty of Health Sciences, Cape Town, South Africa
| | - Elita Avota
- Institute for Virology and Immunobiology, University of Wuerzburg, Versbacher, Wuerzburg, Germany
| | | | - Ayton Meintjes
- Computational Biology Group, Department of Clinical Laboratory Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Faculty of Health Sciences, Cape Town, South Africa
| | - Nicola Mulder
- Computational Biology Group, Department of Clinical Laboratory Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Faculty of Health Sciences, Cape Town, South Africa
| | - Susanne Kneitz
- Department of Physiological Chemistry I, Biocenter, University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| |
Collapse
|
16
|
Wilson EB, Kidani Y, Elsaesser H, Barnard J, Raff L, Karp CL, Bensinger S, Brooks DG. Emergence of distinct multiarmed immunoregulatory antigen-presenting cells during persistent viral infection. Cell Host Microbe 2012; 11:481-91. [PMID: 22607801 DOI: 10.1016/j.chom.2012.03.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 02/10/2012] [Accepted: 03/21/2012] [Indexed: 12/12/2022]
Abstract
During persistent viral infection, adaptive immune responses are suppressed by immunoregulatory factors, contributing to viral persistence. Although this suppression is mediated by inhibitory factors, the mechanisms by which virus-specific T cells encounter and integrate immunoregulatory signals during persistent infection are unclear. We show that a distinct population of IL-10-expressing immunoregulatory antigen-presenting cells (APCs) is amplified during chronic versus acute lymphocytic choriomeningitis virus (LCMV) infection and suppresses T cell responses. Although acute LCMV infection induces the expansion of immunoregulatory APCs, they subsequently decline. However, during persistent LCMV infection, immunoregulatory APCs are amplified and parallel the viral replication kinetics. Further characterization demonstrates that immunoregulatory APCs are molecularly and metabolically distinct, and exhibit increased expression of T cell-interacting molecules and negative regulatory factors that suppress T cell responses. Thus, immunoregulatory APCs are amplified during viral persistence and deliver inhibitory signals that suppress antiviral T cell immunity and likely contribute to persistent infection.
Collapse
Affiliation(s)
- Elizabeth B Wilson
- Department of Microbiology, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Lin WHW, Kouyos RD, Adams RJ, Grenfell BT, Griffin DE. Prolonged persistence of measles virus RNA is characteristic of primary infection dynamics. Proc Natl Acad Sci U S A 2012; 109:14989-94. [PMID: 22872860 PMCID: PMC3443140 DOI: 10.1073/pnas.1211138109] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Measles virus (MeV) is the poster child for acute infection followed by lifelong immunity. However, recent work shows the presence of MeV RNA in multiple sites for up to 3 mo after infection in a proportion of infected children. Here, we use experimental infection of rhesus macaques to show that prolonged RNA presence is characteristic of primary infection. We found that viral RNA persisted in the blood, respiratory tract, or lymph nodes four to five times longer than the infectious virus and that the clearance of MeV RNA from blood happened in three phases: rapid decline coincident with clearance of infectious virus, a rebound phase with increases up to 10-fold, and a phase of slow decrease to undetectable levels. To examine the effect of individual host immune factors on MeV load dynamics further, we developed a mathematical model that expressed viral replication and elimination in terms of the strength of MeV-specific T-cell responses, antibody responses, target cell limitations, and immunosuppressive activity of regulatory T cells. Based on the model, we demonstrate that viral dynamics, although initially regulated by T cells, require antibody to eliminate viral RNA. These results have profound consequences for our view of acute viral infections, the development of prolonged immunity, and, potentially, viral evolution.
Collapse
Affiliation(s)
- Wen-Hsuan W. Lin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205
| | - Roger D. Kouyos
- Department of Ecology and Evolutionary Biology, Woodrow Wilson School of Public and International Affairs, Princeton University, Princeton, NJ 08544
| | - Robert J. Adams
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Bryan T. Grenfell
- Department of Ecology and Evolutionary Biology, Woodrow Wilson School of Public and International Affairs, Princeton University, Princeton, NJ 08544
- Fogarty International Center, National Institutes of Health, Bethesda, MD 20892
| | - Diane E. Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
18
|
Wilson EB, Brooks DG. Translating insights from persistent LCMV infection into anti-HIV immunity. Immunol Res 2011; 48:3-13. [PMID: 20725865 DOI: 10.1007/s12026-010-8162-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human immunodeficiency virus (HIV) is a major global health concern with more than 30 million individuals currently infected worldwide. To date, attempts to stimulate protective immunity to viral components of HIV have been unsuccessful in preventing or clearing infection. Lymphocytic choriomeningitis virus (LCMV) is an established murine model of persistent viral infection that has been instrumental in illuminating several critical aspects of antiviral immunity. Although virologically the course of LCMV infection differs significantly from HIV, the immune responses and regulatory mechanisms elicited by these two viruses are markedly similar. In this review we discuss important recent findings in the LCMV model, highlighting the role of host-derived proteins in shaping immune responses to persistent infections, and explore the therapeutic potential of manipulating these pathways to enhance HIV vaccination strategies.
Collapse
Affiliation(s)
- Elizabeth B Wilson
- Department of Microbiology, Immunology and Molecular Genetics and the UCLA AIDS Institute, David Geffen School of Medicine, University of California-Los Angeles, CA 90095, USA
| | | |
Collapse
|
19
|
Shifting hierarchies of interleukin-10-producing T cell populations in the central nervous system during acute and persistent viral encephalomyelitis. J Virol 2011; 85:6702-13. [PMID: 21525347 DOI: 10.1128/jvi.00200-11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Interleukin-10 (IL-10) mRNA is rapidly upregulated in the central nervous system (CNS) following infection with neurotropic coronavirus and remains elevated during persistent infection. Infection of transgenic IL-10/green fluorescent protein (GFP) reporter mice revealed that CNS-infiltrating T cells were the major source of IL-10, with minimal IL-10 production by macrophages and resident microglia. The proportions of IL-10-producing cells were initially similar in CD8(+) and CD4(+) T cells but diminished rapidly in CD8(+) T cells as the virus was controlled. Overall, the majority of IL-10-producing CD8(+) T cells were specific for the immunodominant major histocompatibility complex (MHC) class I epitope. Unlike CD8(+) T cells, a large proportion of CD4(+) T cells within the CNS retained IL-10 production throughout persistence. Furthermore, elevated frequencies of IL-10-producing CD4(+) T cells in the spinal cord supported preferential maintenance of IL-10 production at the site of viral persistence and tissue damage. IL-10 was produced primarily by the CD25(+) CD4(+) T cell subset during acute infection but prevailed in CD25(-) CD4(+) T cells during the transition to persistent infection and thereafter. Overall, these data demonstrate significant fluidity in the T-cell-mediated IL-10 response during viral encephalitis and persistence. While IL-10 production by CD8(+) T cells was limited primarily to the time of acute effector function, CD4(+) T cells continued to produce IL-10 throughout infection. Moreover, a shift from predominant IL-10 production by CD25(+) CD4(+) T cells to CD25(-) CD4(+) T cells suggests that a transition to nonclassical regulatory T cells precedes and is retained during CNS viral persistence.
Collapse
|
20
|
The role of IL-10 in regulating immunity to persistent viral infections. Curr Top Microbiol Immunol 2010; 350:39-65. [PMID: 20703965 DOI: 10.1007/82_2010_96] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The immune system has evolved multipronged responses that are critical to effectively defend the body from invading pathogens and to clear infection. However, the same weapons employed to eradicate infection can have caustic effects on normal bystander cells. Therefore, tight regulation is vital and the host must balance engendering correct and sufficient immune responses to pathogens while limiting errant and excessive immunopathology. To accomplish this task, a complex network of positive and negative immune signals are delivered, which in most instances successfully eliminate the pathogen. However, in response to some viral infections, immune function is rapidly suppressed leading to viral persistence. Immune suppression is a critical obstacle to the control of many persistent viral infections such as HIV, hepatitis C, and hepatitis B virus, which together affect more than 500 million individuals worldwide. Thus, the ability to therapeutically enhance immunity is a potentially powerful approach to resolve persistent infections. The host-derived cytokine IL-10 is a key player in the establishment and perpetuation of viral persistence. This chapter discusses the role of IL-10 in viral persistence and explores the exciting prospect of therapeutically blocking IL-10 to increase antiviral immunity and vaccine efficacy.
Collapse
|