1
|
Li C, Huynh NPT, Schanz SJ, Windrem MS, Goldman SA. JC virus spread is potentiated by glial replication and demyelination-linked glial proliferation. Brain 2024; 147:4131-4146. [PMID: 39133566 DOI: 10.1093/brain/awae252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 06/02/2024] [Accepted: 06/29/2024] [Indexed: 11/14/2024] Open
Abstract
Progressive multifocal leukoencephalopathy is a demyelinating infection of the immunosuppressed brain, mediated by the gliotropic polyomavirus JCV. JCV replicates in human glial progenitor cells and astrocytes, which undergo viral T-antigen-triggered mitosis, enabling viral replication. We asked whether JCV spread might therefore be accelerated by glial proliferation. Using both in vitro analysis and a human glial chimeric mouse model of JCV infection, we found that dividing human astrocytes supported JCV propagation to a substantially greater degree than did mitotically quiescent cells. Accordingly, bulk and single-cell RNA-sequence analysis revealed that JCV-infected glia differentially manifested cell cycle-linked disruption of both DNA damage response and transcriptional regulatory pathways. In vivo, JCV infection of humanized glial chimeras was greatly accentuated by cuprizone-induced demyelination and its associated mobilization of glial progenitor cells. Importantly, in vivo infection triggered the death of both uninfected and infected glia, reflecting significant bystander death. Together, these data suggest that JCV propagation in progressive multifocal leukoencephalopathy might be accelerated by glial cell division. As such, the accentuated glial proliferation attending disease-associated demyelination might provide an especially favourable environment for JCV propagation, thus potentiating oligodendrocytic bystander death and further accelerating demyelination in susceptible hosts.
Collapse
Affiliation(s)
- Cui Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, China
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester NY 14604, USA
| | - Nguyen P T Huynh
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester NY 14604, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen 1017, Denmark
- Sana Biotechnology, Cambridge, MA 02139, USA
| | - Steven J Schanz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester NY 14604, USA
| | - Martha S Windrem
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester NY 14604, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester NY 14604, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen 1017, Denmark
- Sana Biotechnology, Cambridge, MA 02139, USA
| |
Collapse
|
2
|
de Goër de Herve MG, Dekeyser M, Hendel-Chavez H, Maillart E, Labeyrie C, Adams D, Moreau T, Lubetzki C, Papeix C, Stankoff B, Gasnault J, Taoufik Y. Frequent detection of IFN-gamma -producing memory effector and effector T cells in patients with progressive multifocal leukoencephalopathy. Front Immunol 2024; 15:1416074. [PMID: 39086476 PMCID: PMC11289500 DOI: 10.3389/fimmu.2024.1416074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction Progressive Multifocal Leukoencephalopathy (PML) is a rare and deadly demyelinating disease caused by JC virus (JCV) replication in the central nervous system. PML occurs exclusively in patients with severe underlying immune deficiencies, including AIDS and hematological malignancies. PML has also emerged as a significant threat to patients on potent new immunosuppressive biologics, including natalizumab in multiple sclerosis. Methods Here, we developed an IFN-γ release assay (IGRA) that mainly detects JCV-specific effector memory T cells and effectors T cells in the blood. Results This assay was frequently positive in patients with active PML (with a positive JCV PCR in CSF) of various underlying immunosuppression causes (84% sensitivity). Only 3% of healthy donors had a positive response (97% specificity). The frequency of positivity also increased in multiple sclerosis patients according to the time on natalizumab (up to 36% in patients treated for more than 48 months, who are considered at a higher risk of PML). Discussion The results show this assay's frequent or increased positivity in patients with PML or an increased risk of PML, respectively. The assay may help to stratify the risk of PML.
Collapse
Affiliation(s)
| | - Manon Dekeyser
- INSERM 1186, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Houria Hendel-Chavez
- INSERM 1186, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Elisabeth Maillart
- Department of Neurology, Hôpital Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Céline Labeyrie
- Department of Neurology, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - David Adams
- Department of Neurology, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | | | - Catherine Lubetzki
- Department of Neurology, Hôpital Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Caroline Papeix
- Department of Neurology, Hôpital Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Bruno Stankoff
- Department of Neurology, Hôpital Tenon, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Jacques Gasnault
- INSERM 1186, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Unité de Suite et Réadaptation, Department of Internal Medicine, Hôpital de Bicêtre, Assistance Publique - Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Yassine Taoufik
- INSERM 1186, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
3
|
Butic AB, Spencer SA, Shaheen SK, Lukacher AE. Polyomavirus Wakes Up and Chooses Neurovirulence. Viruses 2023; 15:2112. [PMID: 37896889 PMCID: PMC10612099 DOI: 10.3390/v15102112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
JC polyomavirus (JCPyV) is a human-specific polyomavirus that establishes a silent lifelong infection in multiple peripheral organs, predominantly those of the urinary tract, of immunocompetent individuals. In immunocompromised settings, however, JCPyV can infiltrate the central nervous system (CNS), where it causes several encephalopathies of high morbidity and mortality. JCPyV-induced progressive multifocal leukoencephalopathy (PML), a devastating demyelinating brain disease, was an AIDS-defining illness before antiretroviral therapy that has "reemerged" as a complication of immunomodulating and chemotherapeutic agents. No effective anti-polyomavirus therapeutics are currently available. How depressed immune status sets the stage for JCPyV resurgence in the urinary tract, how the virus evades pre-existing antiviral antibodies to become viremic, and where/how it enters the CNS are incompletely understood. Addressing these questions requires a tractable animal model of JCPyV CNS infection. Although no animal model can replicate all aspects of any human disease, mouse polyomavirus (MuPyV) in mice and JCPyV in humans share key features of peripheral and CNS infection and antiviral immunity. In this review, we discuss the evidence suggesting how JCPyV migrates from the periphery to the CNS, innate and adaptive immune responses to polyomavirus infection, and how the MuPyV-mouse model provides insights into the pathogenesis of JCPyV CNS disease.
Collapse
Affiliation(s)
| | | | | | - Aron E. Lukacher
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA; (A.B.B.); (S.A.S.); (S.K.S.)
| |
Collapse
|
4
|
Gkrania-Klotsas E. Infections in patients with primary immunodeficiencies: Insight into infections in secondary immunodeficiency and transplantation. Transpl Infect Dis 2023; 25:e14124. [PMID: 37573149 DOI: 10.1111/tid.14124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Affiliation(s)
- Effrossyni Gkrania-Klotsas
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
5
|
Mutations in the John Cunningham virus VP1 gene could predispose to the development of progressive multifocal leukoencephalopathy in multiple sclerosis patients undergoing treatment with natalizumab. Mult Scler Relat Disord 2021; 56:103266. [PMID: 34555758 DOI: 10.1016/j.msard.2021.103266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Patients with Multiple Sclerosis (MS) undergoing treatment with natalizumab (NTZ) are at risk of developing progressive multifocal leukoencephalopathy (PML) due to the reactivation of John Cunningham (JC) virus. A relevant characteristic among PML cases is the development of single nucleotide mutations in the VP1 gene of the causal JC virus. The identification of such mutations in timely manner can provide valuable information for MS management. OBJECTIVE To identify mutations along the JC virus VP1 gene in MS patients undergoing treatment with NTZ, and correlate them with anti-JC virus antibody index. METHODS Eighty-eight MS patients, one hundred twenty controls, and six patients with diagnosis of Human Immunodeficiency Virus (HIV) with and without secondary PML were included. JC virus was identified in peripheral blood mononuclear cells and cerebrospinal fluid by PCR. Amplification and sequencing of the entire length of the VP1 gene were performed in all positive clinical samples. RESULTS In MS cases no mutations were observed in the JC virus VP1 gene, but it was positive in HIV controls with PML. Interestingly, the JC virus VP1 gene sequence derived from the HIV patients exhibited a non-silent substitution in position 186 (G → C), leading to an amino acid change (Lys → Asp). We did not find correlation between anti-JC virus antibody index and DNA viral detection. CONCLUSIONS . The identification of single nucleotide mutants in the JC virus VP1 gene might be an early predictive marker to PML for efficient patient treatment and follow-up.
Collapse
|
6
|
de Nattes T, Etienne I, Gerardin E, Bertrand D, Candon S. Lesson for the clinical nephrologist: immune monitoring of human JC-polyomavirus in kidney transplantation. J Nephrol 2021; 35:1901-1905. [PMID: 34487335 DOI: 10.1007/s40620-021-01150-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/22/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Tristan de Nattes
- Nephrology, Kidney Transplant Unit, Rouen University Hospital, 1 rue de Germont, 76031, Rouen, France.
- Department of Immunology, and Biotherapies, Normandy University, UNIROUEN, INSERM, U1234, Rouen University Hospital, 76031, Rouen, France.
| | - Isabelle Etienne
- Nephrology, Kidney Transplant Unit, Rouen University Hospital, 1 rue de Germont, 76031, Rouen, France
| | - Emmanuel Gerardin
- Department of Neuroradiology, Rouen University Hospital, Rouen, France
| | - Dominique Bertrand
- Nephrology, Kidney Transplant Unit, Rouen University Hospital, 1 rue de Germont, 76031, Rouen, France
| | - Sophie Candon
- Department of Immunology, and Biotherapies, Normandy University, UNIROUEN, INSERM, U1234, Rouen University Hospital, 76031, Rouen, France
| |
Collapse
|
7
|
Cortese I, Reich DS, Nath A. Progressive multifocal leukoencephalopathy and the spectrum of JC virus-related disease. Nat Rev Neurol 2020; 17:37-51. [PMID: 33219338 PMCID: PMC7678594 DOI: 10.1038/s41582-020-00427-y] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a devastating CNS infection caused by JC virus (JCV), a polyomavirus that commonly establishes persistent, asymptomatic infection in the general population. Emerging evidence that PML can be ameliorated with novel immunotherapeutic approaches calls for reassessment of PML pathophysiology and clinical course. PML results from JCV reactivation in the setting of impaired cellular immunity, and no antiviral therapies are available, so survival depends on reversal of the underlying immunosuppression. Antiretroviral therapies greatly reduce the risk of HIV-related PML, but many modern treatments for cancers, organ transplantation and chronic inflammatory disease cause immunosuppression that can be difficult to reverse. These treatments — most notably natalizumab for multiple sclerosis — have led to a surge of iatrogenic PML. The spectrum of presentations of JCV-related disease has evolved over time and may challenge current diagnostic criteria. Immunotherapeutic interventions, such as use of checkpoint inhibitors and adoptive T cell transfer, have shown promise but caution is needed in the management of immune reconstitution inflammatory syndrome, an exuberant immune response that can contribute to morbidity and death. Many people who survive PML are left with neurological sequelae and some with persistent, low-level viral replication in the CNS. As the number of people who survive PML increases, this lack of viral clearance could create challenges in the subsequent management of some underlying diseases. In this Review, Cortese et al. provide an overview of the pathobiology and evolving presentations of progressive multifocal leukoencephalopathy and other diseases caused by JC virus, and discuss emerging immunotherapeutic approaches that could increase survival. Progressive multifocal leukoencephalopathy (PML) is a rare, debilitating and often fatal disease of the CNS caused by JC virus (JCV). JCV establishes asymptomatic, lifelong persistent or latent infection in immune competent hosts, but impairment of cellular immunity can lead to reactivation of JCV and PML. PML most commonly occurs in patients with HIV infection or lymphoproliferative disease and in patients who are receiving natalizumab for treatment of multiple sclerosis. The clinical phenotype of PML varies and is shaped primarily by the host immune response; changes in the treatment of underlying diseases associated with PML have changed phenotypes over time. Other clinical manifestations of JCV infection have been described, including granule cell neuronopathy. Survival of PML depends on reversal of the underlying immunosuppression; emerging immunotherapeutic strategies include use of checkpoint inhibitors and adoptive T cell transfer.
Collapse
Affiliation(s)
- Irene Cortese
- Neuroimmunology Clinic, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Avila M, Okai A, Thakolwiboon S, O'Bryan C, Reddy Gopireddy MM, Gorantla S. The effect of tonsillectomy on John Cunningham virus serological status in multiple sclerosis patients: A retrospective case-control study. Mult Scler 2020; 27:1297-1300. [PMID: 33179571 DOI: 10.1177/1352458520971822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tonsils are believed to be the initial site of the John Cunningham virus (JCV) infection. The long-term effect of childhood tonsillectomy on JCV status in multiple sclerosis (MS) patients has not been investigated. In this retrospective case-control study, we analyzed data of 144 JCV seropositive cases and 82 JCV seronegative controls from three outpatient MS clinics in the United States. Early tonsillectomy (before the age of 8) was reported among 8 (5.56%) JCV seropositive subjects and 19 (23.17%) controls. Early tonsillectomy was associated with JCV negative status (adjusted odds ratio = 5.39, 95% confidence interval = 2.13-13.62, p < 0.001) independent of age and gender.
Collapse
Affiliation(s)
- Mirla Avila
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Annette Okai
- Baylor Scott & White Multiple Sclerosis Treatment Center, Baylor University Medical Center, Dallas, TX, USA
| | - Smathorn Thakolwiboon
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Collin O'Bryan
- OSF HealthCare Illinois Neurological Institute and University of Illinois, College of Medicine, Peoria, IL, USA
| | | | - Sasikanth Gorantla
- OSF HealthCare Illinois Neurological Institute and University of Illinois, College of Medicine, Peoria, IL, USA
| |
Collapse
|
9
|
Khoy K, Mariotte D, Defer G, Petit G, Toutirais O, Le Mauff B. Natalizumab in Multiple Sclerosis Treatment: From Biological Effects to Immune Monitoring. Front Immunol 2020; 11:549842. [PMID: 33072089 PMCID: PMC7541830 DOI: 10.3389/fimmu.2020.549842] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis is a chronic demyelinating disease of the central nervous system (CNS) with an autoimmune component. Among the recent disease-modifying treatments available, Natalizumab, a monoclonal antibody directed against the alpha chain of the VLA-4 integrin (CD49d), is a potent inhibitor of cell migration toward the tissues including CNS. It potently reduces relapses and active brain lesions in the relapsing remitting form of the disease. However, it has also been associated with a severe infectious complication, the progressive multifocal leukoencephalitis (PML). Using the standard protocol with an injection every 4 weeks it has been shown by a close monitoring of the drug that trough levels soon reach a plateau with an almost saturation of the target cell receptor as well as a down modulation of this receptor. In this review, mechanisms of action involved in therapeutic efficacy as well as in PML risk will be discussed. Furthermore the interest of a biological monitoring that may be helpful to rapidly adapt treatment is presented. Indeed, development of anti-NAT antibodies, although sometimes unapparent, can be detected indirectly by normalization of CD49d expression on circulating mononuclear cells and might require to switch to another drug. On the other hand a stable modulation of CD49d expression might be useful to follow the circulating NAT levels and apply an extended interval dose scheme that could contribute to limiting the risk of PML.
Collapse
Affiliation(s)
- Kathy Khoy
- Laboratory of Immunology, Department of Biology, CHU Caen Normandie, Caen, France
| | - Delphine Mariotte
- Laboratory of Immunology, Department of Biology, CHU Caen Normandie, Caen, France
| | - Gilles Defer
- Department of Neurology, MS Expert Centre, CHU Caen Normandie, Caen, France.,UMR-S1237, Physiopathology and Imaging of Neurological Disorders, INSERM, Caen, France.,Normandie Université, UNICAEN, Caen, France
| | - Gautier Petit
- Laboratory of Immunology, Department of Biology, CHU Caen Normandie, Caen, France
| | - Olivier Toutirais
- Laboratory of Immunology, Department of Biology, CHU Caen Normandie, Caen, France.,UMR-S1237, Physiopathology and Imaging of Neurological Disorders, INSERM, Caen, France.,Normandie Université, UNICAEN, Caen, France
| | - Brigitte Le Mauff
- Laboratory of Immunology, Department of Biology, CHU Caen Normandie, Caen, France.,UMR-S1237, Physiopathology and Imaging of Neurological Disorders, INSERM, Caen, France.,Normandie Université, UNICAEN, Caen, France
| |
Collapse
|
10
|
Ahye N, Bellizzi A, May D, Wollebo HS. The Role of the JC Virus in Central Nervous System Tumorigenesis. Int J Mol Sci 2020; 21:ijms21176236. [PMID: 32872288 PMCID: PMC7503523 DOI: 10.3390/ijms21176236] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer is the second leading cause of mortality worldwide. The study of DNA tumor-inducing viruses and their oncoproteins as a causative agent in cancer initiation and tumor progression has greatly enhanced our understanding of cancer cell biology. The initiation of oncogenesis is a complex process. Specific gene mutations cause functional changes in the cell that ultimately result in the inability to regulate cell differentiation and proliferation effectively. The human neurotropic Polyomavirus JC (JCV) belongs to the family Polyomaviridae and it is the causative agent of progressive multifocal leukoencephalopathy (PML), which is a fatal neurodegenerative disease in an immunosuppressed state. Sero-epidemiological studies have indicated JCV infection is prevalent in the population (85%) and that initial infection usually occurs during childhood. The JC virus has small circular, double-stranded DNA that includes coding sequences for viral early and late proteins. Persistence of the virus in the brain and other tissues, as well as its potential to transform cells, has made it a subject of study for its role in brain tumor development. Earlier observation of malignant astrocytes and oligodendrocytes in PML, as well as glioblastoma formation in non-human primates inoculated with JCV, led to the hypothesis that JCV plays a role in central nervous system (CNS) tumorigenesis. Some studies have reported the presence of both JC viral DNA and its proteins in several primary brain tumor specimens. The discovery of new Polyomaviruses such as the Merkel cell Polyomavirus, which is associated with Merkel cell carcinomas in humans, ignited our interest in the role of the JC virus in CNS tumors. The current evidence known about JCV and its effects, which are sufficient to produce tumors in animal models, suggest it can be a causative factor in central nervous system tumorigenesis. However, there is no clear association between JCV presence in CNS and its ability to initiate CNS cancer and tumor formation in humans. In this review, we will discuss the correlation between JCV and tumorigenesis of CNS in animal models, and we will give an overview of the current evidence for the JC virus’s role in brain tumor formation.
Collapse
|
11
|
Early reduction of the splicing factor2/alternative splicing factor: a cellular inhibitor of the JC polyomavirus in natalizumab-treated MS patients long before developing progressive multifocal leukoencephalopathy. J Neurovirol 2019; 26:133-137. [PMID: 31468472 DOI: 10.1007/s13365-019-00793-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/03/2019] [Accepted: 08/14/2019] [Indexed: 10/26/2022]
Abstract
Natalizumab is effective against relapsing-remitting multiple sclerosis (MS) but increases the risk of progressive multifocal leukoencephalopathy (PML), which is caused by the activation of the JCV polyomavirus. SF2/ASF (splicing factor2/alternative splicing factor) is a potent cellular inhibitor of JCV replication and large T-antigen (T-Ag) expression. We reported that SF2/ASF levels in blood cells increase during the first year of natalizumab therapy and decrease thereafter, inversely related to T-Ag expression, and suggested a correlation with JCV reactivation. Here, we report SF2/ASF levels of longitudinal blood samples of two patients undergoing natalizumab therapy, who developed PML while monitored, in comparison to natalizumab-treated controls and to one-off PML samples. After 6 months of therapy, SF2/ASF levels of the two cases were reduced, instead of increased, and their overall SF2/ASF levels were lower than those from natalizumab controls. Since SF2/ASF inhibits JCV, its early reduction might have a role in subsequent PML. We are aware of the limitations of the study, but the uniqueness of serial blood samples collected before and after PML onset in natalizumab-treated patients must be stressed. If confirmed in other patients, SF2/ASF evaluation could be a new and early biomarker of natalizumab-associated PML risk, allowing an 18-24-month interval before PML onset (presently ~ 5 months), in which clinicians could evaluate other risk factors and change therapy.
Collapse
|
12
|
Sandhu MR, Rutledge R, Grant M, Mahajan A, Spudich S. Slowly progressive fatal PML-IRIS following antiretroviral initiation at CD4+ nadir of 350 cells/mm 3 despite CD4+ cell count rise to 900 cells/mm 3. Int J STD AIDS 2019; 30:810-813. [PMID: 31046614 DOI: 10.1177/0956462419835966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
AIDS-related progressive multifocal leukoencephalopathy (PML)-immune reconstitution inflammatory syndrome (IRIS) is a central nervous system inflammatory syndrome where immune response to John Cunningham (JC) virus antigen following antiretroviral therapy (ART) causes breakdown of the blood–brain barrier. We report a unique case of PML-IRIS, which presented with dystonic choreoathetosis after initiation of ART at a CD4+ cell count of 350 cells/mm3. This report shows continuous progression of the disease over a period of two years, despite robust immune reconstitution. The worsening of neurological symptoms, persistent positivity of JC virus in CSF, and progressive inflammatory picture on MR scans in the setting of a CD4+ cell count of 900 cells/mm3 highlight a different variant of PML-IRIS, and challenge the role of CD4+ cell count in diagnosing opportunistic infections in HIV/AIDS patients.
Collapse
Affiliation(s)
- Mani Ratnesh Sandhu
- 1 Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ronnye Rutledge
- 2 Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Matthew Grant
- 3 Department of Internal Medicine, Section of Infectious Disease, Yale School of Medicine, New Haven, CT, USA
| | - Amit Mahajan
- 4 Department of Radiology and Biomedical Imaging, Section of Neuroradiology, Yale School of Medicine, New Haven, CT, USA
| | - Serena Spudich
- 5 Department of Neurology, Division of Neurological Infections & Global Neurology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
13
|
Corbridge SM, Rice RC, Bean LA, Wüthrich C, Dang X, Nicholson DA, Koralnik IJ. JC virus infection of meningeal and choroid plexus cells in patients with progressive multifocal leukoencephalopathy. J Neurovirol 2019; 25:520-524. [PMID: 31025264 DOI: 10.1007/s13365-019-00753-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/07/2019] [Accepted: 04/08/2019] [Indexed: 01/18/2023]
Abstract
JC virus (JCV) can cause a lytic infection of oligodendrocytes and astrocytes in the central nervous system (CNS) leading to progressive multifocal leukoencephalopathy (PML). JCV can also infect meningeal and choroid plexus cells causing JCV meningitis (JCVM). Whether JCV also infects meningeal and choroid plexus cells in PML patients and other immunosuppressed individuals with no overt symptoms of meningitis remains unknown. We therefore analyzed archival formalin-fixed, paraffin-embedded brain samples from PML patients, and HIV-seropositive and seronegative control subjects by immunohistochemistry for the presence of JCV early regulatory T Ag and JCV VP1 late capsid protein. In meninges, we detected JCV T Ag in 11/48 (22.9%) and JCV VP1 protein in 8/48 (16.7%) PML patients. In choroid plexi, we detected JCV T Ag in 1/7 (14.2%) and JCV VP1 protein in 1/8 (12.5%) PML patients. Neither JCV T Ag nor VP1 protein could be detected in meninges or choroid plexus of HIV-seropositive and HIV-seronegative control subjects without PML. In addition, examination of underlying cerebellar cortex of PML patients revealed JCV-infected cells in the molecular layer, including GAD 67+ interneurons, but not in HIV-seropositive and HIV-seronegative control subjects without PML. Our findings suggest that productive JCV infection of meningeal cells and choroid plexus cells also occurs in PML patients without signs or symptoms of meningitis. The phenotypic characterization of JCV-infected neurons in the molecular layer deserves further study. This data provides new insight into JCV pathogenesis in the CNS.
Collapse
Affiliation(s)
- Sarah M Corbridge
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Richard C Rice
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Linda A Bean
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Christian Wüthrich
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Boston, MA, USA
| | - Xin Dang
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Daniel A Nicholson
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Igor J Koralnik
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
14
|
Tiwari S, Lapierre J, Ojha CR, Martins K, Parira T, Dutta RK, Caobi A, Garbinski L, Ceyhan Y, Esteban-Lopez M, El-Hage N. Signaling pathways and therapeutic perspectives related to environmental factors associated with multiple sclerosis. J Neurosci Res 2018; 96:1831-1846. [PMID: 30204260 PMCID: PMC7167107 DOI: 10.1002/jnr.24322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disorder of unknown etiology. Both genetic-susceptibility and environment exposures, including vitamin D deficiency, Epstein-Barr viral and Herpesvirus (HHV-6) infections are strongly implicated in the activation of T cells and MS-pathogenesis. Despite precise knowledge of how these factors could be operating alone or in combination to facilitate and aggravate the disease progression, it is clear that prolonged induction of inflammatory molecules and recruitment of other immune cells by the activated T cells results in demyelination and axonal damage. It is imperative to understand the risk factors associated with MS progression and how these factors contribute to disease pathology. Understanding of the underlying mechanisms of what factors triggers activation of T cells to attack myelin antigen are important to strategize therapeutics and therapies against MS. Current review provides a detailed literature to understand the role of both pathogenic and non-pathogenic factors on the impact of MS.
Collapse
Affiliation(s)
- Sneham Tiwari
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Jessica Lapierre
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Chet Raj Ojha
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Kyle Martins
- Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Tiyash Parira
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Rajib Kumar Dutta
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Allen Caobi
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Luis Garbinski
- Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Yasemin Ceyhan
- Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Maria Esteban-Lopez
- Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Nazira El-Hage
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| |
Collapse
|
15
|
Jaureguiberry-Bravo M, Lopez L, Berman JW. Frontline Science: Buprenorphine decreases CCL2-mediated migration of CD14 + CD16 + monocytes. J Leukoc Biol 2018; 104:1049-1059. [PMID: 29791013 DOI: 10.1002/jlb.3hi0118-015r] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/26/2018] [Accepted: 04/23/2018] [Indexed: 12/28/2022] Open
Abstract
HIV infection of the CNS causes neuroinflammation and damage that contributes to the development of HIV-associated neurocognitive disorders (HAND) in greater than 50% of HIV-infected individuals, despite antiretroviral therapy (ART). Opioid abuse is a major risk factor for HIV infection. It has been shown that opioids can contribute to increased HIV CNS pathogenesis, in part, by modulating the function of immune cells. HIV enters the CNS within two weeks after peripheral infection by transmigration of infected monocytes across the blood brain barrier (BBB). CD14+ CD16+ monocytes are a mature subpopulation that is increased in number in the peripheral blood of HIV-infected people. Mature monocytes can be productively infected with HIV, and they transmigrate preferentially across the BBB in response to CCL2, a chemokine elevated in the CNS and CSF of HIV-infected people even with ART. Buprenorphine, an opioid derivate, is an opioid replacement therapy for heroin addiction. It is a partial agonist of μ-opioid receptor and full antagonist of κ-opioid receptor. The effects of buprenorphine on CCL2-mediated CD14+ CD16+ monocytes transmigration across the BBB, a critical mechanism that promotes neuroinflammation and HAND, have not been characterized. We showed for the first time that buprenorphine decreases several steps of CCL2-mediated human mature monocyte transmigration. We propose that buprenorphine treatment in the context of HIV infection could serve a dual purpose, to treat opioid addiction and also to reduce neuroinflammation. Additionally, buprenorphine may be used as a treatment for HAND not only in the context of opioid abuse.
Collapse
Affiliation(s)
| | - Lillie Lopez
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Joan W Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
16
|
Pathogenesis of progressive multifocal leukoencephalopathy and risks associated with treatments for multiple sclerosis: a decade of lessons learned. Lancet Neurol 2018; 17:467-480. [DOI: 10.1016/s1474-4422(18)30040-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 11/30/2017] [Accepted: 01/25/2018] [Indexed: 12/12/2022]
|
17
|
Abstract
Multiple sclerosis (MS) has long been considered a CD4 T-cell disease, primarily because of the findings that the strongest genetic risk for MS is the major histocompatibility complex (MHC) class II locus, and that T cells play a central role in directing the immune response. The importance that the T helper (Th)1 cytokine, interferon γ (IFN-γ), and the Th17 cytokine, interleukin (IL)-17, play in MS pathogenesis is indicated by recent clinical trial data by the enhanced presence of Th1/Th17 cells in central nervous system (CNS) tissue, cerebrospinal fluid (CSF), and blood, and by research on animal models of MS, such as experimental autoimmune encephalomyelitis (EAE). Although the majority of research on MS pathogenesis has centered on the role of effector CD4 T cells, accumulating data suggests that CD8 T cells may play a significant role in the human disease. In fact, in contrast to most animal models, the primary T cell found in the CNS in patients with MS, is the CD8 T cell. As patient-derived effector T cells are also resistant to mechanisms of dominant tolerance such as that induced by interaction with regulatory T cells (Tregs), their reduced response to regulation may also contribute to the unchecked effector T-cell activity in patients with MS. These concepts will be discussed below.
Collapse
Affiliation(s)
- Belinda J Kaskow
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Clare Baecher-Allan
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
18
|
T cell deficiencies as a common risk factor for drug associated progressive multifocal leukoencephalopathy. Immunobiology 2018; 223:508-517. [PMID: 29472141 DOI: 10.1016/j.imbio.2018.01.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/19/2018] [Accepted: 01/21/2018] [Indexed: 01/07/2023]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a disease of the central nervous system caused by neuropathogenic prototypes of ubiquitous community-acquired JC virus (JCV). The disease became of particular concern following its association with certain therapies that modulate immune system function without heavy immunosuppression. Due to lack of prophylactic/treatment options and poor outcomes, which often include severe disability or death, PML is a considerable concern for development of new drugs that interfere with immune system functions. In this review of clinical and research findings, we discuss the evidence that deficiencies in CD4+ T helper cells, cytotoxic CD8+ T cells, and interferon gamma are of crucial importance for development of PML under a variety of circumstances, including those associated with use of various drugs, regardless of differences in their mechanisms of action. These deficiencies apparently enable transformation of the harmless JCV archetype into neuropathogenic prototypes, but the site(s), and the mechanisms, of this transformation are yet to be elucidated. Here we discuss the evidence for brain as one of the sites of this transformation, and propose a model of PML pathogenesis that emphasizes the central role of T cell deficiencies in the two life cycles of the JCV, one non-pathogenic and one neuropathogenic. Finally, we conclude that the development of clinical grade T cell functional tests and more consistent use of already available laboratory tests for T cell subset analysis would greatly aid the effort to more accurately predict and assess the magnitude of PML risk for concerned therapeutic interventions.
Collapse
|
19
|
Hu C, Huang Y, Su J, Wang M, Zhou Q, Zhu B. Detection and analysis of variants of JC polyomavirus in urine samples from HIV-1-infected patients in China's Zhejiang Province. J Int Med Res 2018; 46:1024-1032. [PMID: 29322824 PMCID: PMC5972266 DOI: 10.1177/0300060517746297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Objectives Human JC polyomavirus (JCPyV) infection has an increased risk of developing progressive multifocal leukoencephalopathy (PML). Different JCPyV subtypes differ in the virulence with which they cause PML. Currently, the JCPyV infection status and subtype distribution in patients with human immunodeficiency virus-1 (HIV-1) in China are still unclear. This study aimed to investigate the epidemiology and subtype distribution of JCPyV in HIV-1-infected patients in China. Methods Urine samples from 137 HIV-1-infected patients in Zhejiang Province in China were tested for the presence of JCPyV DNA. The detected VP1 sequences were aligned and analysed using BioEdit and MEGA software. Results Among urine samples from HIV-1-infected patients, 67.2% were positive for JCPyV DNA (92/137). Primarily, the type 7 strains of JCPyV were detected, among which 45.5% (15/33) were subtype 7A, 30.3% (10/33) were 7B, and 24.2% (8/33) were 7C. Six nucleotide mutations, as well as one amino acid substitution, were isolated from the patients. Conclusions Urine samples from HIV-1-infected patients from Zhejiang Province show a high JCPyV infection rate. The most common JCPyV strains are subtypes 7A, 7B, and 7C.
Collapse
Affiliation(s)
- Caiqin Hu
- Department of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, 71069 College of Medicine, Zhejiang University , Hangzhou, China
| | - Ying Huang
- Department of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, 71069 College of Medicine, Zhejiang University , Hangzhou, China
| | - Junwei Su
- Department of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, 71069 College of Medicine, Zhejiang University , Hangzhou, China
| | - Mengyan Wang
- Department of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, 71069 College of Medicine, Zhejiang University , Hangzhou, China
| | - Qihui Zhou
- Department of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, 71069 College of Medicine, Zhejiang University , Hangzhou, China
| | - Biao Zhu
- Department of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, 71069 College of Medicine, Zhejiang University , Hangzhou, China
| |
Collapse
|
20
|
Protective Humoral Immunity in the Central Nervous System Requires Peripheral CD19-Dependent Germinal Center Formation following Coronavirus Encephalomyelitis. J Virol 2017; 91:JVI.01352-17. [PMID: 28931676 DOI: 10.1128/jvi.01352-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023] Open
Abstract
B cell subsets with phenotypes characteristic of naive, non-isotype-switched, memory (Bmem) cells and antibody-secreting cells (ASC) accumulate in various models of central nervous system (CNS) inflammation, including viral encephalomyelitis. During neurotropic coronavirus JHMV infection, infiltration of protective ASC occurs after T cell-mediated viral control and is preceded by accumulation of non-isotype-switched IgD+ and IgM+ B cells. However, the contribution of peripheral activation events in cervical lymph nodes (CLN) to driving humoral immune responses in the infected CNS is poorly defined. CD19, a signaling component of the B cell receptor complex, is one of multiple regulators driving B cell differentiation and germinal center (GC) formation by lowering the threshold of antigen-driven activation. JHMV-infected CD19-/- mice were thus used to determine how CD19 affects CNS recruitment of B cell subsets. Early polyclonal ASC expansion, GC formation, and virus-specific ASC were all significantly impaired in CLN of CD19-/- mice compared to wild-type (WT) mice, consistent with lower and unsustained virus-specific serum antibody (Ab). ASC were also significantly reduced in the CNS, resulting in increased infectious virus during persistence. Nevertheless, CD19 deficiency did not affect early CNS IgD+ B cell accumulation. The results support the notion that CD19-independent factors drive early B cell mobilization and recruitment to the infected CNS, while delayed accumulation of virus-specific, isotype-switched ASC requires CD19-dependent GC formation in CLN. CD19 is thus essential for both sustained serum Ab and protective local Ab within the CNS following JHMV encephalomyelitis.IMPORTANCE CD19 activation is known to promote GC formation and to sustain serum Ab responses following antigen immunization and viral infections. However, the contribution of CD19 in the context of CNS infections has not been evaluated. This study demonstrates that antiviral protective ASC in the CNS are dependent on CD19 activation and peripheral GC formation, while accumulation of early-recruited IgD+ B cells is CD19 independent. This indicates that IgD+ B cells commonly found early in the CNS do not give rise to local ASC differentiation and that only antigen-primed, peripheral GC-derived ASC infiltrate the CNS, thereby limiting potentially harmful nonspecific Ab secretion. Expanding our understanding of activation signals driving CNS migration of distinct B cell subsets during neuroinflammatory insults is critical for preventing and managing acute encephalitic infections, as well as preempting reactivation of persistent viruses during immune-suppressive therapies targeting B cells in multiple sclerosis (MS), such as rituximab and ocrelizumab.
Collapse
|
21
|
Cerebellar Involvement in an Immunocompetent Patient Presenting with Progressive Multifocal Leukoencephalopathy. Case Rep Neurol Med 2017; 2017:2396068. [PMID: 28912987 PMCID: PMC5587930 DOI: 10.1155/2017/2396068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 08/02/2017] [Indexed: 11/17/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is a demyelinating disease caused by the JC virus, a polyomavirus that can be reactivated under certain immunosuppressive conditions, such as AIDS, immunomodulatory therapy, and haematological malignancies. However, a few cases of immunocompetent patients have been reported in which no immunodeficiency was present. We describe the case of an 83-year-old immunocompetent man who presented with severe cerebellar symptoms with an MRI scan suggestive of severe demyelinating disease. We were not able to identify any occult immunosuppression or malignancy in our patient.
Collapse
|
22
|
Misbah SA. Progressive multi-focal leucoencephalopathy - driven from rarity to clinical mainstream by iatrogenic immunodeficiency. Clin Exp Immunol 2017; 188:342-352. [PMID: 28245526 PMCID: PMC5422720 DOI: 10.1111/cei.12948] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2017] [Indexed: 12/21/2022] Open
Abstract
Advances in immune-mediated targeted therapies have proved to be a double-edged sword for patients by highlighting the risk of iatrogenic infective complications. This has been exemplified by progressive multi-focal leucoencephalopathy (PML), a hitherto rare devastating viral infection of the brain caused by the neurotrophic JC polyoma virus. While PML achieved prominence during the first two decades of the HIV epidemic, effective anti-retroviral treatment and restitution of T cell function has led to PML being less prominent in this population. HIV infection as a predisposing factor has now been supplanted by T cell immunodeficiency induced by a range of immune-mediated therapies as a major cause of PML. This review focuses on PML in the context of therapeutic immunosuppression and encompasses therapeutic monoclonal antibodies, novel immunomodulatory agents such as Fingolimod and dimethyl fumarate, as well as emerging data on PML in primary immune deficiency.
Collapse
Affiliation(s)
- S A Misbah
- Department of Clinical Immunology, Oxford University Hospitals, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
23
|
Uleri E, Ibba G, Piu C, Caocci M, Leoni S, Arru G, Serra C, Sechi G, Dolei A. JC polyomavirus expression and bell-shaped regulation of its SF2/ASF suppressor during the follow-up of multiple sclerosis patients treated with natalizumab. J Neurovirol 2016; 23:226-238. [PMID: 27812788 DOI: 10.1007/s13365-016-0492-x] [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: 07/08/2016] [Revised: 09/19/2016] [Accepted: 10/10/2016] [Indexed: 12/14/2022]
Abstract
Natalizumab is effective against multiple sclerosis (MS), but is associated with progressive multifocal leukoencephalopathy (PML), fatal disease caused by the JCV polyomavirus. The SF2/ASF (splicing factor2/alternative splicing factor) inhibits JCV in glial cells. We wondered about SF2/ASF modulation in the blood of natalizumab-treated patients and if this could influence JCV reactivation. Therefore, we performed a longitudinal study of MS patients under natalizumab, in comparison to patients under fingolimod and to healthy blood donors. Blood samples were collected at time intervals. The expression of SF2/ASF and the presence and expression of JCV in PBMC were analyzed. A bell-shaped regulation of SF2/ASF was observed in patients treated with natalizumab, increased in the first year of therapy, and reduced in the second one, while slightly changed, if any, in patients under fingolimod. Notably, SF2/ASF was up-regulated, during the first year, only in JCV DNA-positive patients, or with high anti-JCV antibody response; the expression of the JCV T-Ag protein in circulating B cells was inversely related to SF2/ASF protein expression. The SF2/ASF reduction, parallel to JCV activation, during the second year of therapy with natalizumab, but not with fingolimod, may help explain the increased risk of PML after the second year of treatment with natalizumab, but not with fingolimod. We propose that SF2/ASF has a protective role against JCV reactivation in MS patients. This study suggests new markers of disease behavior and, possibly, help in re-evaluations of therapy protocols.
Collapse
Affiliation(s)
- Elena Uleri
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, I-07100, Sassari, Italy
| | - Gabriele Ibba
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, I-07100, Sassari, Italy
| | - Claudia Piu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, I-07100, Sassari, Italy
| | - Maurizio Caocci
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, I-07100, Sassari, Italy
| | - Stefania Leoni
- Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 8, I-07100, Sassari, Italy
| | - Giannina Arru
- Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 8, I-07100, Sassari, Italy
| | - Caterina Serra
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, I-07100, Sassari, Italy
| | - GianPietro Sechi
- Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 8, I-07100, Sassari, Italy
| | - Antonina Dolei
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, I-07100, Sassari, Italy.
| |
Collapse
|
24
|
Saylor D, Venkatesan A. Progressive Multifocal Leukoencephalopathy in HIV-Uninfected Individuals. Curr Infect Dis Rep 2016; 18:33. [PMID: 27686675 DOI: 10.1007/s11908-016-0543-8] [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] [Indexed: 12/20/2022]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a demyelinating disease of the central nervous system (CNS) caused by the human neurotropic polyomavirus JC (JCV). The disease occurs virtually exclusively in immunocompromised individuals, and, prior to the introduction of antiretroviral therapy, was seen most commonly in the setting of HIV/AIDS. More recently, however, the incidence of PML in HIV-uninfected persons has increased with broader use of immunosuppressive and immunomodulatory medications utilized in a variety of systemic and neurologic autoimmune disorders. In this review, we discuss the epidemiology and clinical characteristics of PML in HIV-uninfected individuals, as well as diagnostic modalities and the limited treatment options. Moreover, we describe recent findings regarding the neuropathogenesis of PML, with specific focus on the unique association between PML and natalizumab, a monoclonal antibody that prevents trafficking of activated leukocytes into the CNS that is used for the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Deanna Saylor
- Division of Neuroimmunology and Neuro-Infectious Diseases, Department of Neurology, The Johns Hopkins University School of Medicine, Meyer 6-113, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Arun Venkatesan
- Division of Neuroimmunology and Neuro-Infectious Diseases, Department of Neurology, The Johns Hopkins University School of Medicine, Meyer 6-113, 600 N. Wolfe Street, Baltimore, MD, 21287, USA.
| |
Collapse
|
25
|
Lepetit M, Laplaud DA. Effetti collaterali delle bioterapie nella sclerosi multipla e nelle malattie correlate. Neurologia 2016. [DOI: 10.1016/s1634-7072(16)78802-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
26
|
Zerbe CS, Marciano BE, Katial RK, Santos CB, Adamo N, Hsu AP, Hanks ME, Darnell DN, Quezado MM, Frein C, Barnhart LA, Anderson VL, Uzel G, Freeman AF, Lisco A, Nath A, Major EO, Sampaio EP, Holland SM. Progressive Multifocal Leukoencephalopathy in Primary Immune Deficiencies: Stat1 Gain of Function and Review of the Literature. Clin Infect Dis 2016; 62:986-94. [PMID: 26743090 PMCID: PMC4803104 DOI: 10.1093/cid/civ1220] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 12/04/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Progressive multifocal leukoencephalopathy (PML) is a rare, severe, otherwise fatal viral infection of the white matter of the brain caused by the polyomavirus JC virus, which typically occurs only in immunocompromised patients. One patient with dominant gain-of-function (GOF) mutation in signal transducer and activator of transcription 1 (STAT1) with chronic mucocutaneous candidiasis and PML was reported previously. We aim to identify the molecular defect in 3 patients with PML and to review the literature on PML in primary immune defects (PIDs). METHODS STAT1 was sequenced in 3 patients with PML. U3C cell lines were transfected with STAT1 and assays to search for STAT1 phosphorylation, transcriptional response, and target gene expression were performed. RESULTS We identified 3 new unrelated cases of PML in patients with GOF STAT1 mutations, including the novel STAT1 mutation, L400Q. These STAT1 mutations caused delayed STAT1 dephosphorylation and enhanced interferon-gamma-driven responses. In our review of the literature regarding PML in primary immune deficiencies we found 26 cases, only 54% of which were molecularly characterized, the remainder being syndromically diagnosed only. CONCLUSIONS The occurrence of PML in 4 cases of STAT1 GOF suggests that STAT1 plays a critical role in the control of JC virus in the central nervous system.
Collapse
Affiliation(s)
- Christa S Zerbe
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Beatriz E Marciano
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Rohit K Katial
- National Jewish Health and University of Colorado, Health Sciences Center, Denver
| | - Carah B Santos
- National Jewish Health and University of Colorado, Health Sciences Center, Denver
| | - Nick Adamo
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Amy P Hsu
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Mary E Hanks
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Dirk N Darnell
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Martha M Quezado
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda
| | - Cathleen Frein
- Clinical Research Directorate/Clinical Monitoring Research Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick
| | - Lisa A Barnhart
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Victoria L Anderson
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Gulbu Uzel
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Alexandra F Freeman
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Andrea Lisco
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | | | - Eugene O Major
- Laboratory of Molecular Medicine and Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth P Sampaio
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Steven M Holland
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
27
|
Domínguez-Mozo MI, Toledano-Martínez E, Rodríguez-Rodríguez L, García-Montojo M, Alvarez-Lafuente R, Fernández-Gutiérrez B. JC virus reactivation in patients with autoimmune rheumatic diseases treated with rituximab. Scand J Rheumatol 2016; 45:507-511. [DOI: 10.3109/03009742.2015.1135980] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- MI Domínguez-Mozo
- Departments of Neurology and Rheumatology, Health Research Institute, San Carlos Clinical Hospital (IDISSC), Madrid, Spain
| | - E Toledano-Martínez
- Departments of Neurology and Rheumatology, Health Research Institute, San Carlos Clinical Hospital (IDISSC), Madrid, Spain
| | - L Rodríguez-Rodríguez
- Departments of Neurology and Rheumatology, Health Research Institute, San Carlos Clinical Hospital (IDISSC), Madrid, Spain
| | - M García-Montojo
- Departments of Neurology and Rheumatology, Health Research Institute, San Carlos Clinical Hospital (IDISSC), Madrid, Spain
| | - R Alvarez-Lafuente
- Departments of Neurology and Rheumatology, Health Research Institute, San Carlos Clinical Hospital (IDISSC), Madrid, Spain
| | - B Fernández-Gutiérrez
- Departments of Neurology and Rheumatology, Health Research Institute, San Carlos Clinical Hospital (IDISSC), Madrid, Spain
| |
Collapse
|
28
|
|
29
|
Brief Report: Role of Thymic Reconstitution in the Outcome of AIDS-Related PML. J Acquir Immune Defic Syndr 2016; 70:357-61. [PMID: 26181821 DOI: 10.1097/qai.0000000000000754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Implications of thymopoiesis in AIDS-related opportunistic infections remain unexplored. We used progressive multifocal leukoencephalopathy (PML), caused by JC virus (JCV), as an opportunistic infection model, and we simultaneously investigated thymic output and T-cell responses against JCV in 22 patients with PML treated with combined antiretroviral therapy. Thymic output was significantly associated with JCV-specific CD4⁺ and CD8⁺ T-cell responses and improved survival. Our data suggest that patients with AIDS-related PML and impaired thymopoiesis are less likely to develop a robust JCV-specific cellular immune response and consequently are at an increased risk for a poor clinical outcome.
Collapse
|
30
|
Kampylafka EI, Alexopoulos H, Dalakas MC, Tzioufas AG. Immunotherapies for Neurological Manifestations in the Context of Systemic Autoimmunity. Neurotherapeutics 2016; 13:163-78. [PMID: 26510559 PMCID: PMC4720664 DOI: 10.1007/s13311-015-0393-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Neurological involvement is relatively common in the majority of systemic autoimmune diseases and may lead to severe morbidity and mortality, if not promptly treated. Treatment options vary greatly, depending on the underlying systemic pathophysiology and the associated neurological symptoms. Selecting the appropriate therapeutic scheme is further complicated by the lack of definite therapeutic guidelines, the necessity to differentiate primary neurological syndromes from those related to the underlying systemic disease, and to sort out adverse neurological manifestations caused by immunosuppressants or the biological agents used to treat the primary disease. Immunotherapy is a sine qua non for treating most, if not all, neurological conditions presenting in the context of systemic autoimmunity. Specific agents include classical immune modulators such as corticosteroids, cyclophosphamide, intravenous immunoglobulin, and plasma exchange, as well as numerous biological therapies, for example anti-tumor necrosis factor agents and monoclonal antibodies that target various immune pathways such as B cells, cytokines, and co-stimulatory molecules. However, experience regarding the use of these agents in neurological complications of systemic diseases is mainly empirical or based on small uncontrolled studies and case series. The aim of this review is to present the state-of-the-art therapies applied in various neurological manifestations encountered in the context of systemic autoimmune diseases; evaluate all treatment options on the basis of existing guidelines; and compliment these data with our personal experience derived from a large number of patients.
Collapse
Affiliation(s)
- Eleni I Kampylafka
- Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Harry Alexopoulos
- Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Marinos C Dalakas
- Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Athanasios G Tzioufas
- Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece.
| |
Collapse
|
31
|
Bellaguarda E, Keyashian K, Pekow J, Rubin DT, Cohen RD, Sakuraba A. Prevalence of Antibodies Against JC Virus in Patients With Refractory Crohn's Disease and Effects of Natalizumab Therapy. Clin Gastroenterol Hepatol 2015; 13:1919-25. [PMID: 26001336 PMCID: PMC4795937 DOI: 10.1016/j.cgh.2015.05.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/21/2015] [Accepted: 05/05/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Natalizumab, a humanized antibody against the α4 integrin subunit, effectively induces and maintains remission in patients with Crohn's disease (CD) refractory to conventional treatments. Progressive multifocal leukoencephalopathy is a rare but fatal brain infection caused by John Cunningham (JC) virus and has been associated with natalizumab use. We assessed the prevalence of and risk factors for antibodies to JC virus in serum of patients with refractory CD who were candidates for, or already were receiving, natalizumab. We also assessed the effects of natalizumab treatment of these patients. METHODS In a retrospective study, we analyzed clinical charts from 191 patients with CD (74 males; mean age, 38.7 y; mean duration of disease, 14.9 y) tested for serum JC virus antibody from December 2012 through May 2014 at 2 medical centers in the United States. We calculated JC virus antibody prevalence and compared the characteristics of patients who tested negative vs those who tested positive, to identify risk factors. We also assessed the rate of subsequent natalizumab use, surgery, and seroconversion during natalizumab therapy. RESULTS A total of 129 of the patients (67.5%) tested positive for serum JC virus antibody. Multivariate analysis showed that past use of thiopurine was a risk factor for testing positive for JC virus antibody (odds ratio, 7.8; 95% confidence interval, 2.0-30.4; P = .003). Twenty-two of the patients who tested negative for JC virus antibody (35.5%) and 16 of the 129 patients who tested positive (12.4%) had been treated with natalizumab. Cox regression analysis determined that natalizumab use was the only factor associated with avoiding subsequent surgery (hazard ratio, 0.23; 95% confidence interval, 0.06-0.98). Seroconversion (from testing negative to positive for JC virus antibody) occurred in 1 of the 22 patients (4.5%) who initially tested negative during natalizumab therapy. CONCLUSIONS The prevalence of CD patients exposed to JC virus is comparable with that of the general population. In this retrospective study, prior thiopurine use was associated with an increased risk for testing positive for JC virus antibody. Natalizumab use reduced the risk of subsequent surgery.
Collapse
Affiliation(s)
- Emanuelle Bellaguarda
- Inflammatory Bowel Disease Center, The University of Chicago Medicine, Chicago, Illinois
| | - Kian Keyashian
- Division of Gastroenterology and Hepatology, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Joel Pekow
- Inflammatory Bowel Disease Center, The University of Chicago Medicine, Chicago, Illinois
| | - David T Rubin
- Inflammatory Bowel Disease Center, The University of Chicago Medicine, Chicago, Illinois
| | - Russell D Cohen
- Inflammatory Bowel Disease Center, The University of Chicago Medicine, Chicago, Illinois
| | - Atsushi Sakuraba
- Inflammatory Bowel Disease Center, The University of Chicago Medicine, Chicago, Illinois.
| |
Collapse
|
32
|
Domínguez-Mozo MI, García-Montojo M, Arias-Leal A, García-Martínez Á, Santiago JL, Casanova I, Galán V, Arroyo R, Fernández-Arquero M, Alvarez-Lafuente R. Monitoring the John Cunningham virus throughout natalizumab treatment in multiple sclerosis patients. Eur J Neurol 2015; 23:182-9. [DOI: 10.1111/ene.12834] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 08/04/2015] [Indexed: 12/21/2022]
Affiliation(s)
- M. I. Domínguez-Mozo
- Department of Neurology; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Hospital Clínico San Carlos; Madrid Spain
| | - M. García-Montojo
- Department of Neurology; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Hospital Clínico San Carlos; Madrid Spain
| | - A. Arias-Leal
- Department of Neurology; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Hospital Clínico San Carlos; Madrid Spain
| | - Á. García-Martínez
- Department of Neurology; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Hospital Clínico San Carlos; Madrid Spain
| | - J. L. Santiago
- Department of Immunology; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Hospital Clínico San Carlos; Madrid Spain
| | - I. Casanova
- Department of Neurology; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Hospital Clínico San Carlos; Madrid Spain
| | - V. Galán
- Department of Neurology; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Hospital Clínico San Carlos; Madrid Spain
| | - R. Arroyo
- Department of Neurology; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Hospital Clínico San Carlos; Madrid Spain
| | - M. Fernández-Arquero
- Department of Immunology; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Hospital Clínico San Carlos; Madrid Spain
| | - R. Alvarez-Lafuente
- Department of Neurology; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Hospital Clínico San Carlos; Madrid Spain
| |
Collapse
|
33
|
Lack of Major Histocompatibility Complex Class I Upregulation and Restrictive Infection by JC Virus Hamper Detection of Neurons by T Lymphocytes in the Central Nervous System. J Neuropathol Exp Neurol 2015; 74:791-803. [PMID: 26115192 DOI: 10.1097/nen.0000000000000218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The human polyomavirus JC (JCV) infects glial cells in immunosuppressed individuals, leading to progressive multifocal leukoencephalopathy. Polyomavirus JC can also infect neurons in patients with JCV granule cell neuronopathy and JCV encephalopathy. CD8-positive T cells play a crucial role in viral containment and outcome in progressive multifocal leukoencephalopathy, but whether CD8-positive T cells can also recognize JCV-infected neurons is unclear. We used immunohistochemistry to determine the prevalence of T cells in neuron-rich areas of archival brain samples from 77 patients with JCV CNS infections and 94 control subjects. Neurons predominantly sustained a restrictive infection with expression of JCV regulatory protein T antigen (T Ag), whereas glial cells were productively infected and expressed both T Ag and the capsid protein VP1. T cells were more prevalent near JCV-infected cells with intact nuclei expressing both T Ag and VP1 compared with those expressing either protein alone. CD8-positive T cells also colocalized more with JCV-infected glial cells than with JCV-infected neurons. Major histocompatibility complex class I expression was upregulated in JCV-infected areas but could only be detected in rare neurons interspersed with infected glial cells. These results suggest that isolated neurons harboring restrictive JCV infection do not upregulate major histocompatibility complex class I and thus may escape recognition by CD8-positive T cells.
Collapse
|
34
|
Dallari S, Franciotta D, Carluccio S, Signorini L, Gastaldi M, Colombo E, Bergamaschi R, Elia F, Villani S, Ferrante P, Delbue S. Upregulation of integrin expression on monocytes in multiple sclerosis patients treated with natalizumab. J Neuroimmunol 2015; 287:76-9. [PMID: 26439965 DOI: 10.1016/j.jneuroim.2015.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/28/2015] [Accepted: 08/12/2015] [Indexed: 11/15/2022]
Abstract
Natalizumab is a humanized monoclonal antibody against the α4 subunit of VLA-4 integrin that is used to treat conditions such as multiple sclerosis (MS). Although its effects on lymphocytes have been widely described, little is known about its effects on monocytes. Here we described the effects of natalizumab treatment on peripheral blood monocytes from a small cohort of MS patients in terms of relative frequencies and surface integrin (CD49d and CD18) expression. We showed that natalizumab treatment altered the surface integrin expression on monocyte subsets in the peripheral compartment, suggesting a role for them as mediators of natalizumab effects.
Collapse
Affiliation(s)
- Simone Dallari
- Department of Biomedical, Surgical and Dental Sciences, University of Milano, Milano, Italy
| | - Diego Franciotta
- Department of General Neurology, National Neurological Institute C. Mondino, Pavia, Italy
| | - Silvia Carluccio
- Department of Biomedical, Surgical and Dental Sciences, University of Milano, Milano, Italy
| | - Lucia Signorini
- Department of Biomedical, Surgical and Dental Sciences, University of Milano, Milano, Italy
| | - Matteo Gastaldi
- Department of General Neurology, National Neurological Institute C. Mondino, Pavia, Italy
| | - Elena Colombo
- Department of General Neurology, National Neurological Institute C. Mondino, Pavia, Italy
| | - Roberto Bergamaschi
- Department of General Neurology, National Neurological Institute C. Mondino, Pavia, Italy
| | - Francesca Elia
- Department of Biomedical, Surgical and Dental Sciences, University of Milano, Milano, Italy
| | - Sonia Villani
- Department of Biomedical, Surgical and Dental Sciences, University of Milano, Milano, Italy
| | - Pasquale Ferrante
- Department of Biomedical, Surgical and Dental Sciences, University of Milano, Milano, Italy; Fondazione Ettore Sansavini, Health Science Foundation, Lugo, Italy
| | - Serena Delbue
- Department of Biomedical, Surgical and Dental Sciences, University of Milano, Milano, Italy.
| |
Collapse
|
35
|
Monaco MCG, Major EO. Immune System Involvement in the Pathogenesis of JC Virus Induced PML: What is Learned from Studies of Patients with Underlying Diseases and Therapies as Risk Factors. Front Immunol 2015; 6:159. [PMID: 25972864 PMCID: PMC4412132 DOI: 10.3389/fimmu.2015.00159] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/24/2015] [Indexed: 12/16/2022] Open
Abstract
The human polyomavirus JC PyV lytic infection of oligodendrocytes in the human brain results in the demyelinating disease progressive multifocal leukoencephalopathy, PML. JCV is a common virus infection in the population that leads to PML in patients with underlying diseases and therapies that cause immune deficiencies or modulate immune system functions. Patients may have high levels of antibody to JCV that neither protect them from PML nor clear the infection once PML is established. Cell-mediated immunity plays a more effective role in clearing initial or reactivated JCV infection before PML occurs. However, patients with underlying diseases and therapies for treatment are at high risk for PML. MS patients on natalizumab are one of the categories with the highest incidence of PML. Natalizumab is a humanized monoclonal antibody targeting α4 integrins that prevents inflammatory cells from entering the brain and it has been used as a treatment for MS. A number of studies have investigated the occurrence of PML in these patients and their cell-mediated immune profile that might gain insight into the mechanism that ties natalizumab with a high risk of developing PML. It seems that cells of the immune system participate in the pathogenesis of PML as well as clearance of JCV infection.
Collapse
Affiliation(s)
- Maria Chiara G Monaco
- Laboratory of Molecular Medicine and Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health , Bethesda, MD , USA
| | - Eugene O Major
- Laboratory of Molecular Medicine and Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
36
|
Mattoscio M, Nicholas R, Sormani MP, Malik O, Lee JS, Waldman AD, Dazzi F, Muraro PA. Hematopoietic mobilization: Potential biomarker of response to natalizumab in multiple sclerosis. Neurology 2015; 84:1473-82. [PMID: 25762712 DOI: 10.1212/wnl.0000000000001454] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/22/2014] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To ascertain the mobilization from the bone marrow and the functional relevance of the increased number of circulating hematopoietic stem and progenitor cells (HSPC) induced by the anti-α-4 integrin antibody natalizumab in patients with multiple sclerosis (MS). METHODS We evaluated CD45(low)CD34+ HSPC frequency by flow cytometry in blood from 45 natalizumab-treated patients (12 of whom were prospectively followed during the first year of treatment as part of a pilot cohort and 16 prospectively followed for validation), 10 untreated patients with MS, and 24 healthy donors. In the natalizumab-treated group, we also assessed sorted HSPC cell cycle status, T- and B-lymphocyte subpopulation frequencies (n = 29), and HSPC differentiation potential (n = 10). RESULTS Natalizumab-induced circulating HSPC were predominantly quiescent, suggesting recent mobilization from the bone marrow, and were capable of differentiating ex vivo. Circulating HSPC numbers were significantly increased during natalizumab, but heterogeneously, allowing the stratification of mobilizer and nonmobilizer subgroups. Nonmobilizer status was associated with persistence of disease activity during treatment. The frequency of B cells and CD103+CD8+ regulatory T cells persistently increased, more significantly in mobilizer patients, who also showed a specific naive/memory B-cell profile. CONCLUSIONS The data suggest that natalizumab-induced circulating HSPC increase is the result of true mobilization from the bone marrow and has clinical and immunologic relevance. HSPC mobilization, associated with clinical remission and increased proportion of circulating B and regulatory T cells, may contribute to the treatment's mode of action; thus, HSPC blood counts could represent an early biomarker of responsiveness to natalizumab.
Collapse
Affiliation(s)
- Miriam Mattoscio
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Richard Nicholas
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Maria P Sormani
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Omar Malik
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Jean S Lee
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Adam D Waldman
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Francesco Dazzi
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Paolo A Muraro
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy.
| |
Collapse
|
37
|
Wollebo HS, White MK, Gordon J, Berger JR, Khalili K. Persistence and pathogenesis of the neurotropic polyomavirus JC. Ann Neurol 2015; 77:560-70. [PMID: 25623836 DOI: 10.1002/ana.24371] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/23/2014] [Accepted: 01/08/2015] [Indexed: 12/11/2022]
Abstract
Many neurological diseases of the central nervous system (CNS) are underpinned by malfunctions of the immune system, including disorders involving opportunistic infections. Progressive multifocal leukoencephalopathy (PML) is a lethal CNS demyelinating disease caused by the human neurotropic polyomavirus JC (JCV) and is found almost exclusively in individuals with immune disruption, including patients with human immunodeficiency virus/acquired immunodeficiency syndrome, patients receiving therapeutic immunomodulatory monoclonal antibodies to treat conditions such as multiple sclerosis, and transplant recipients. Thus, the public health significance of this disease is high, because of the number of individuals constituting the at-risk population. The incidence of PML is very low, whereas seroprevalence for the virus is high, suggesting infection by the virus is very common, and so it is thought that the virus is restrained but it persists in an asymptomatic state that can only occasionally be disrupted to lead to viral reactivation and PML. When JCV actively replicates in oligodendrocytes and astrocytes of the CNS, it produces cytolysis, leading to formation of demyelinated lesions with devastating consequences. Defining the molecular nature of persistence and events leading to reactivation of the virus to cause PML has proved to be elusive. In this review, we examine the current state of knowledge of the JCV life cycle and mechanisms of pathogenesis. We will discuss the normal course of the JCV life cycle including transmission, primary infection, viremia, and establishment of asymptomatic persistence as well as pathogenic events including migration of the virus to the brain, reactivation from persistence, viral infection, and replication in the glial cells of the CNS and escape from immunosurveillance.
Collapse
Affiliation(s)
- Hassen S Wollebo
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA
| | | | | | | | | |
Collapse
|
38
|
Jelcic I, Jelcic I, Faigle W, Sospedra M, Martin R. Immunology of progressive multifocal leukoencephalopathy. J Neurovirol 2015; 21:614-22. [PMID: 25740538 DOI: 10.1007/s13365-014-0294-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/10/2014] [Accepted: 09/18/2014] [Indexed: 11/30/2022]
Abstract
The high prevalence of asymptomatic JC polyomavirus (JCV) infection in the general population indicates coexistence with the human host and efficient immune control in healthy individuals. For unknown reasons, kidney-resident archetypic JCV strains can turn into neurotropic JCV strains which in hereditary or acquired states of immunodeficiency cause opportunistic infection and cytolytic destruction of glial cells or granule cell neurons resulting in progressive multifocal demyelination in the central nervous system (CNS) or cerebellar atrophy, respectively. Immunomodulatory or immunosuppressive therapies with specific monoclonal antibodies including natalizumab, efalizumab, and rituximab have increased the risk of progressive multifocal leukoencephalopathy (PML) among treated patients, highlighting that symptomatic JCV infection of the CNS is associated with disturbances of adaptive immunity affecting B cells, antibodies, and CD4(+) and/or CD8(+) T cells. To date, no specific therapy to overcome PML is available and the only way to eliminate the virus from the CNS is to reconstitute global immune function. However, since the identification of JCV as the causative agent of PML 40 years ago, it is still not fully understood which components of the immune system prevent the development of PML and which immune mechanisms are involved in eliminating the virus from the CNS. This review gives an update about adaptive JCV-specific immune responses.
Collapse
Affiliation(s)
- Ivan Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Ilijas Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Wolfgang Faigle
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, 8091, Zurich, Switzerland.
| |
Collapse
|
39
|
Molecular diagnostic tests to predict the risk of progressive multifocal leukoencephalopathy in natalizumab-treated multiple sclerosis patients. Mol Cell Probes 2015; 29:54-62. [DOI: 10.1016/j.mcp.2014.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 11/26/2014] [Accepted: 11/26/2014] [Indexed: 11/18/2022]
|
40
|
Antoniol C, Stankoff B. Immunological Markers for PML Prediction in MS Patients Treated with Natalizumab. Front Immunol 2015; 5:668. [PMID: 25601865 PMCID: PMC4283628 DOI: 10.3389/fimmu.2014.00668] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/11/2014] [Indexed: 12/15/2022] Open
Abstract
Natalizumab (NTZ), a monoclonal antibody recognizing the alpha4 integrin chain, has been approved for the treatment of active multiple sclerosis, but expose to the onset of a rare side effect, progressive multifocal leukoencephalopathy (PML). Estimating the individual risk of PML in NTZ-treated patients is a major challenge, and therapeutic strategies are mainly guided by the overall PML risk assessed by identified risk factors: JC virus (JCV) seropositivity, treatment duration (with peak incidence after 24 months), and the previous use of immunosuppressive therapies. Given that this stratification does not yet allow a precise individual prediction of PML, other predictive markers are needed, and several immunological biomarkers have been described. Quantification of anti-JCV antibody levels may improve individual predictive value, with higher baseline titers indicating increased risk. Other immunological biomarkers such as leukocyte cell membrane markers (CD49d, CD11a, and CD62L), detection of circulating JCV-specific activated T effector memory cells (TEM) or genetic screening have been proposed. In this review, we discuss how recent progress in immunology has paved the way for «new combined monitoring», which will include immunological screening, in NTZ-treated patients.
Collapse
Affiliation(s)
- Caroline Antoniol
- AP-HP, Hôpital Saint-Antoine , Paris , France ; Centre Hospitalier Universitaire de Dijon, Université de Bourgogne , Dijon , France
| | - Bruno Stankoff
- AP-HP, Hôpital Saint-Antoine , Paris , France ; Sorbonne Universités, Université Pierre et Marie Curie, UMR S 1127, CNRS UMR 7225, and l'Institut du Cerveau et de la Moelle Épinière (ICM) , Paris , France
| |
Collapse
|
41
|
Warnke C, Stettner M, Lehmensiek V, Dehmel T, Mausberg AK, von Geldern G, Gold R, Kümpfel T, Hohlfeld R, Mäurer M, Stangel M, Straeten V, Limmroth V, Weber T, Kleinschnitz C, Wattjes MP, Svenningsson A, Olsson T, Hartung HP, Hermsen D, Tumani H, Adams O, Kieseier BC. Natalizumab exerts a suppressive effect on surrogates of B cell function in blood and CSF. Mult Scler 2014; 21:1036-44. [PMID: 25392339 DOI: 10.1177/1352458514556296] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 09/08/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND Natalizumab for multiple sclerosis (MS) increases the risk of progressive multifocal leukoencephalopathy (PML). OBJECTIVE We aimed to assess the effect of natalizumab on cellular composition and functional B cell parameters including patients with natalizumab-associated PML (n=37). METHODS Cellular composition by flow cytometry, levels of immunoglobulin (Ig)G/IgM by immunonephelometry, and oligoclonal bands by isoelectric focusing were studied in blood and cerebrospinal fluid. RESULTS In MS patients treated with natalizumab without PML (n=59) the proportion of CD19+ B cells was higher in blood, but lower in cerebrospinal fluid compared with MS patients not treated with natalizumab (n=17). The CD4/CD8-ratio in cerebrospinal fluid was lower, and IgG and IgM levels as well as the IgG index dropped in longitudinal samples during natalizumab therapy. Oligoclonal bands persisted, but the total amount of the intrathecally produced IgG fraction, and the polyclonal intrathecal IgG reactivity to measles, rubella, and zoster declined. At the time of diagnosis of PML patients with natalizumab-associated PML had low total IgG levels in blood and cerebrospinal fluid. CONCLUSIONS Natalizumab impacts B and T cell distribution and exerts an inhibitory effect on surrogates of B cell function in periphery and in cerebrospinal fluid, potentially contributing to the increased risk of developing PML.
Collapse
Affiliation(s)
- Clemens Warnke
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Mark Stettner
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | | | - Thomas Dehmel
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Anne K Mausberg
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Gloria von Geldern
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Ralf Gold
- Department of Neurology, Ruhr University, Bochum, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, Ludwig Maximilian University and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, Ludwig Maximilian University and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mathias Mäurer
- Department of Neurology, Caritas Hospital, Bad Mergentheim, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hanover Medical School, Hanover, Germany
| | - Vera Straeten
- Department of Neurology, Johannes Wesling Hospital Minden, Minden, Germany
| | | | - Thomas Weber
- Department of Neurology, Marienhospital Hamburg, Hamburg, Germany
| | | | - Mike P Wattjes
- MS Center Amsterdam and Department of Radiology, Nuclear Medicine & PET Research, VU University Medical Center, Amsterdam, the Netherlands
| | - Anders Svenningsson
- Department of Pharmacology and Clinical Neuroscience, Umea University Hospital, Umea, Sweden
| | - Tomas Olsson
- Department of Neurology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Derik Hermsen
- Institute for Clinical Chemistry and Laboratory Diagnostics, University Hospital Duesseldorf, Germany
| | | | - Ortwin Adams
- Institute for Virology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Bernd C Kieseier
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| |
Collapse
|
42
|
Münz C. Viral infections in mice with reconstituted human immune system components. Immunol Lett 2014; 161:118-24. [PMID: 24953718 DOI: 10.1016/j.imlet.2014.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/14/2014] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
Abstract
Pathogenic viruses are often difficult to study due to their exclusive tropism for humans. The development of mice with human immune system components opens the possibility to study those human pathogens with a tropism for the human hematopoietic lineage in vivo. These include HCMV, EBV, KSHV, HIV, HTLV-1, dengue virus and JC virus. Furthermore, some human pathogens, like HSV-2, adenovirus, HCV, HBV and influenza A virus, with an additional tropism for somatic mouse tissues or for additional transplanted human tissues, mainly liver, have been explored in these models. The cellular tropism of these viruses, their associated diseases and primarily cell-mediated immune responses to these viral infections will be discussed in this review. Already some exciting information has been gained from these novel chimeric in vivo models and future avenues to gain more insights into the pathology, but also potential therapies, will be outlined. Although the respective in vivo models of human immune responses can still be significantly improved, they already provide preclinical systems for in vivo studies of important viral pathogens of humans.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Switzerland.
| |
Collapse
|