Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.725
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 36: 667-694, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677479

ABSTRACT

Pattern recognition receptors (PRRs) survey intra- and extracellular spaces for pathogen-associated molecular patterns (PAMPs) within microbial products of infection. Recognition and binding to cognate PAMP ligand by specific PRRs initiates signaling cascades that culminate in a coordinated intracellular innate immune response designed to control infection. In particular, our immune system has evolved specialized PRRs to discriminate viral nucleic acid from host. These are critical sensors of viral RNA to trigger innate immunity in the vertebrate host. Different families of PRRs of virus infection have been defined and reveal a diversity of PAMP specificity for wide viral pathogen coverage to recognize and extinguish virus infection. In this review, we discuss recent insights in pathogen recognition by the RIG-I-like receptors, related RNA helicases, Toll-like receptors, and other RNA sensor PRRs, to present emerging themes in innate immune signaling during virus infection.


Subject(s)
DEAD Box Protein 58/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Virus Diseases/etiology , Virus Diseases/metabolism , Viruses/immunology , Animals , DEAD-box RNA Helicases/metabolism , Humans , Protein Processing, Post-Translational , RNA Helicases/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Receptors, Immunologic , Signal Transduction , Toll-Like Receptors/metabolism
2.
Nat Immunol ; 21(12): 1563-1573, 2020 12.
Article in English | MEDLINE | ID: mdl-33106669

ABSTRACT

Chronic cytomegalovirus (CMV) infection leads to long-term maintenance of extraordinarily large CMV-specific T cell populations. The magnitude of this so-called 'memory inflation' is thought to mainly depend on antigenic stimulation during the chronic phase of infection. However, by mapping the long-term development of CD8+ T cell families derived from single naive precursors, we find that fate decisions made during the acute phase of murine CMV infection can alter the level of memory inflation by more than 1,000-fold. Counterintuitively, a T cell family's capacity for memory inflation is not determined by its initial expansion. Instead, those rare T cell families that dominate the chronic phase of infection show an early transcriptomic signature akin to that of established T central memory cells. Accordingly, a T cell family's long-term dominance is best predicted by its early content of T central memory precursors, which later serve as a stem-cell-like source for memory inflation.


Subject(s)
Clonal Evolution/immunology , Host-Pathogen Interactions/immunology , Immunologic Memory , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Virus Diseases/etiology , Virus Diseases/metabolism , Acute Disease , Animals , Biomarkers , Chronic Disease , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Gene Expression Profiling , Humans , Immunophenotyping , Mice , Muromegalovirus/immunology
3.
Blood ; 141(8): 877-885, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36574622

ABSTRACT

Adoptively transferred virus-specific T cells (VSTs) have shown remarkable safety and efficacy for the treatment of virus-associated diseases and malignancies in hematopoietic stem cell transplant (HSCT) recipients, for whom VSTs are derived from the HSCT donor. Autologous VSTs have also shown promise for the treatment of virus-driven malignancies outside the HSCT setting. In both cases, VSTs are manufactured as patient-specific products, and the time required for procurement, manufacture, and release testing precludes their use in acutely ill patients. Further, Good Manufacturing Practices-compliant products are expensive, and failures are common in virus-naive HSCT donors and patient-derived VSTs that are rendered anergic by immunosuppressive tumors. Hence, highly characterized, banked VSTs (B-VSTs) that can be used for multiple unrelated recipients are highly desirable. The major challenges facing B-VSTs result from the inevitable mismatches in the highly polymorphic and immunogenic human leukocyte antigens (HLA) that present internally processed antigens to the T-cell receptor, leading to the requirement for partial HLA matching between the B-VST and recipient. HLA mismatches lead to rapid rejection of allogeneic T-cell products and graft-versus-host disease induced by alloreactive T cells in the infusion product. Here, we summarize the clinical outcomes to date of trials of B-VSTs used for the treatment of viral infections and malignancies and their potential as a platform for chimeric antigen receptors targeting nonviral tumors. We will highlight the properties of VSTs that make them attractive off-the-shelf cell therapies, as well as the challenges that must be overcome before they can become mainstream.


Subject(s)
Hematopoietic Stem Cell Transplantation , Virus Diseases , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , T-Lymphocytes , Cell- and Tissue-Based Therapy , Virus Diseases/etiology , Receptors, Antigen, T-Cell , HLA Antigens
4.
Blood ; 141(17): 2062-2074, 2023 04 27.
Article in English | MEDLINE | ID: mdl-36493341

ABSTRACT

Preventing viral infections at an early stage is a key strategy for successfully improving transplant outcomes. Preemptive therapy and prophylaxis with antiviral agents have been successfully used to prevent clinically significant viral infections in hematopoietic cell transplant recipients. Major progress has been made over the past decades in preventing viral infections through a better understanding of the biology and risk factors, as well as the introduction of novel antiviral agents and advances in immunotherapy. High-quality evidence exists for the effective prevention of herpes simplex virus, varicella-zoster virus, and cytomegalovirus infection and disease. Few data are available on the effective prevention of human herpesvirus 6, Epstein-Barr virus, adenovirus, and BK virus infections. To highlight the spectrum of clinical practice, here we review high-risk situations that we handle with a high degree of uniformity and cases that feature differences in approaches, reflecting distinct hematopoietic cell transplant practices, such as ex vivo T-cell depletion.


Subject(s)
Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Virus Diseases , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/prevention & control , Herpesvirus 4, Human , Virus Diseases/prevention & control , Virus Diseases/etiology , Antiviral Agents/therapeutic use
5.
PLoS Biol ; 19(4): e3001135, 2021 04.
Article in English | MEDLINE | ID: mdl-33878111

ABSTRACT

Identifying the animal reservoirs from which zoonotic viruses will likely emerge is central to understanding the determinants of disease emergence. Accordingly, there has been an increase in studies attempting zoonotic "risk assessment." Herein, we demonstrate that the virological data on which these analyses are conducted are incomplete, biased, and rapidly changing with ongoing virus discovery. Together, these shortcomings suggest that attempts to assess zoonotic risk using available virological data are likely to be inaccurate and largely only identify those host taxa that have been studied most extensively. We suggest that virus surveillance at the human-animal interface may be more productive.


Subject(s)
Environmental Monitoring , Virus Diseases , Zoonoses/etiology , Zoonoses/prevention & control , Animals , Biodiversity , Disease Reservoirs/classification , Disease Reservoirs/statistics & numerical data , Environmental Monitoring/methods , Environmental Monitoring/standards , Host Specificity/genetics , Humans , Metagenomics/methods , Metagenomics/organization & administration , Metagenomics/standards , Phylogeny , Risk Assessment , Risk Factors , Selection Bias , Virus Diseases/epidemiology , Virus Diseases/etiology , Virus Diseases/prevention & control , Virus Diseases/transmission , Viruses/classification , Viruses/genetics , Viruses/isolation & purification , Viruses/pathogenicity , Zoonoses/epidemiology , Zoonoses/virology
6.
Am J Hematol ; 99(4): 662-678, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38197307

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy, an innovative immunotherapeutic against relapsed/refractory B-cell lymphoma, faces challenges due to frequent viral infections. Despite this, a comprehensive review addressing risk assessment, surveillance, and treatment management is notably absent. This review elucidates immune response compromises during viral infections in CAR-T recipients, collates susceptibility risk factors, and deliberates on preventive strategies. In the post-pandemic era, marked by the Omicron variant, new and severe threats to CAR-T therapy emerge, necessitating exploration of preventive and treatment measures for COVID-19. Overall, the review provides recommendations for viral infection prophylaxis and management, enhancing CAR-T product safety and recipient survival.


Subject(s)
Lymphoma, B-Cell , Receptors, Chimeric Antigen , Virus Diseases , Humans , Receptors, Chimeric Antigen/therapeutic use , Immunotherapy, Adoptive/adverse effects , Lymphoma, B-Cell/therapy , Virus Diseases/etiology , Antigens, CD19 , Cell- and Tissue-Based Therapy
7.
J Allergy Clin Immunol ; 151(6): 1634-1645, 2023 06.
Article in English | MEDLINE | ID: mdl-36638922

ABSTRACT

BACKGROUND: Allogenic hematopoietic stem cell transplantation (HSCT) and gene therapy (GT) are potentially curative treatments for severe combined immunodeficiency (SCID). Late-onset posttreatment manifestations (such as persistent hepatitis) are not uncommon. OBJECTIVE: We sought to characterize the prevalence and pathophysiology of persistent hepatitis in transplanted SCID patients (SCIDH+) and to evaluate risk factors and treatments. METHODS: We used various techniques (including pathology assessments, metagenomics, single-cell transcriptomics, and cytometry by time of flight) to perform an in-depth study of different tissues from patients in the SCIDH+ group and corresponding asymptomatic similarly transplanted SCID patients without hepatitis (SCIDH-). RESULTS: Eleven patients developed persistent hepatitis (median of 6 years after HSCT or GT). This condition was associated with the chronic detection of enteric viruses (human Aichi virus, norovirus, and sapovirus) in liver and/or stools, which were not found in stools from the SCIDH- group (n = 12). Multiomics analysis identified an expansion of effector memory CD8+ T cells with high type I and II interferon signatures. Hepatitis was associated with absence of myeloablation during conditioning, split chimerism, and defective B-cell function, representing 25% of the 44 patients with SCID having these characteristics. Partially myeloablative retransplantation or GT of patients with this condition (which we have named as "enteric virus infection associated with hepatitis") led to the reconstitution of T- and B-cell immunity and remission of hepatitis in 5 patients, concomitantly with viral clearance. CONCLUSIONS: Enteric virus infection associated with hepatitis is related to chronic enteric viral infection and immune dysregulation and is an important risk for transplanted SCID patients with defective B-cell function.


Subject(s)
Enterovirus Infections , Hematopoietic Stem Cell Transplantation , Hepatitis , Severe Combined Immunodeficiency , Virus Diseases , Humans , Severe Combined Immunodeficiency/therapy , Severe Combined Immunodeficiency/etiology , CD8-Positive T-Lymphocytes , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Virus Diseases/etiology , Hepatitis/etiology
8.
Curr Opin Infect Dis ; 36(6): 529-536, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37729657

ABSTRACT

PURPOSE OF REVIEW: Seasonal respiratory virus infections (RVIs) often progress to severe diseases in hematopoietic cell transplant (HCT) recipients. This review summarizes the current evidence on risk factors for the severity of RVIs in this high-risk population and provides clinical management. RECENT FINDINGS: The likelihood of the respiratory viral disease progression depends on the immune status of the host and the type of virus. Conventional host factors, such as the immunodeficiency scoring index and the severe immunodeficiency criteria, have been utilized to estimate the risk of progression to severe disease, including mortality. Recent reports have suggested nonconventional risk factors, such as hyperglycemia, hypoalbuminemia, prior use of antibiotics with broad anaerobic activity, posttransplant cyclophosphamide, and pulmonary impairment after RVIs. Identifying novel and modifiable risk factors is important with the advances of novel therapeutic and preventive interventions for RVIs. SUMMARY: Validation of recently identified risk factors for severe RVIs in HCT recipients is required. The development of innovative interventions along with appropriate risk stratification is critical to improve outcomes in this vulnerable population.


Subject(s)
Hematopoietic Stem Cell Transplantation , Respiratory Tract Infections , Virus Diseases , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Transplant Recipients , Seasons , Risk Factors , Virus Diseases/epidemiology , Virus Diseases/etiology , Respiratory Tract Infections/epidemiology , Retrospective Studies
9.
PLoS Pathog ; 17(5): e1009555, 2021 05.
Article in English | MEDLINE | ID: mdl-34015063

ABSTRACT

Although a growing number of studies suggest interactions between Schistosoma parasites and viral infections, the effects of schistosome infections on the host response to viruses have not been evaluated comprehensively. In this systematic review, we investigated how schistosomes impact incidence, virulence, and prevention of viral infections in humans and animals. We also evaluated immune effects of schistosomes in those coinfected with viruses. We screened 4,730 studies and included 103. Schistosomes may increase susceptibility to some viruses, including HIV and Kaposi's sarcoma-associated herpesvirus, and virulence of hepatitis B and C viruses. In contrast, schistosome infection may be protective in chronic HIV, Human T-cell Lymphotropic Virus-Type 1, and respiratory viruses, though further research is needed. Schistosome infections were consistently reported to impair immune responses to hepatitis B and possibly measles vaccines. Understanding the interplay between schistosomes and viruses has ramifications for anti-viral vaccination strategies and global control of viral infections.


Subject(s)
Antiviral Agents/pharmacology , Coinfection/prevention & control , Immunity/immunology , Schistosoma/immunology , Schistosomiasis/complications , Virus Diseases/prevention & control , Viruses/immunology , Animals , Coinfection/etiology , Coinfection/pathology , Humans , Schistosomiasis/parasitology , Virus Diseases/etiology , Virus Diseases/pathology
10.
Haematologica ; 108(8): 2080-2090, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36794500

ABSTRACT

Therapy-resistant viral reactivations contribute significantly to mortality after hematopoietic stem cell transplantation. Adoptive cellular therapy with virus-specific T cells (VST) has shown efficacy in various single-center trials. However, the scalability of this therapy is hampered by laborious production methods. In this study we describe the in-house production of VST in a closed system (CliniMACS Prodigy® system, Miltenyi Biotec). In addition, we report the efficacy in 26 patients with viral disease following hematopoietic stem cell transplantation in a retrospective analysis (adenovirus, n=7; cytomegalovirus, n=8; Epstein-Barr virus, n=4; multi-viral, n=7). The production of VST was successful in 100% of cases. The safety profile of VST therapy was favorable (n=2 grade 3 and n=1 grade 4 adverse events; all three were reversible). A response was seen in 20 of 26 patients (77%). Responding patients had a significantly better overall survival than patients who did not respond (P<0.001). Virus-specific symptoms were reduced or resolved in 47% of patients. The overall survival of the whole cohort was 28% after 6 months. This study shows the feasibility of automated VST production and safety of application. The scalability of the CliniMACS Prodigy® device increases the accessibility of VST treatment.


Subject(s)
Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Virus Diseases , Humans , T-Lymphocytes , Epstein-Barr Virus Infections/therapy , Retrospective Studies , Herpesvirus 4, Human , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Virus Diseases/etiology , Virus Diseases/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Stem Cell Transplantation
11.
Infect Immun ; 90(1): e0032121, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34606367

ABSTRACT

The gut microbiota has emerged as a critical player in host health. Bacteroides fragilis is a prominent member of the gut microbiota within the phyla Bacteroidetes. This commensal bacterium produces unique capsular polysaccharides processed by antigen-presenting cells and activates CD4+ T cells to secrete inflammatory cytokines. Indeed, due to their immunomodulatory functions, B. fragilis and its capsular polysaccharide-A (PSA) are arguably the most explored single commensal microbiota/symbiotic factor. B. fragilis/PSA has been shown to protect against colitis, encephalomyelitis, colorectal cancer, pulmonary inflammation, and asthma. Here, we review recent data on the immunomodulatory role of B. fragilis/PSA during viral infections and therapy, B. fragilis PSA's dual ability to mediate pro-and anti-inflammatory processes, and the potential for exploring this unique characteristic during intracellular bacterial infections such as with Mycobacterium tuberculosis. We also discuss the protective roles of single commensal-derived probiotic species, including B. fragilis in lung inflammation and respiratory infections that may provide essential cues for possible exploration of microbiota based/augmented therapies in tuberculosis (TB). Available data on the relationship between B. fragilis/PSA, the immune system, and disease suggest clinical relevance for developing B. fragilis into a next-generation probiotic or, possibly, the engineering of PSA into a potent carbohydrate-based vaccine.


Subject(s)
Bacteroides fragilis/physiology , Gastrointestinal Microbiome , Host-Pathogen Interactions , Microbial Interactions , Virus Diseases/etiology , Virus Diseases/therapy , Antibiosis , Cytokines/metabolism , Disease Management , Disease Resistance/immunology , Disease Susceptibility , Host-Pathogen Interactions/immunology , Humans , Immunomodulation , Inflammation Mediators/metabolism , Interferons/metabolism , Organ Specificity , Polysaccharides, Bacterial/immunology , Probiotics , Symbiosis , Tuberculosis/etiology , Virus Diseases/metabolism
12.
Eur J Immunol ; 51(5): 1039-1061, 2021 05.
Article in English | MEDLINE | ID: mdl-33729549

ABSTRACT

Type I IFNs are so-named because they interfere with viral infection in vertebrate cells. The study of cellular responses to type I IFNs led to the discovery of the JAK-STAT signaling pathway, which also governs the response to other cytokine families. We review here the outcome of viral infections in mice and humans with engineered and inborn deficiencies, respectively, of (i) IFNAR1 or IFNAR2, selectively disrupting responses to type I IFNs, (ii) STAT1, STAT2, and IRF9, also impairing cellular responses to type II (for STAT1) and/or III (for STAT1, STAT2, IRF9) IFNs, and (iii) JAK1 and TYK2, also impairing cellular responses to cytokines other than IFNs. A picture is emerging of greater redundancy of human type I IFNs for protective immunity to viruses in natural conditions than was initially anticipated. Mouse type I IFNs are essential for protection against a broad range of viruses in experimental conditions. These findings suggest that various type I IFN-independent mechanisms of human cell-intrinsic immunity to viruses have yet to be discovered.


Subject(s)
Genetic Predisposition to Disease , Interferon Type I/metabolism , Signal Transduction , Virus Diseases/etiology , Virus Diseases/metabolism , Alleles , Animals , Disease Models, Animal , Genotype , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/deficiency , Interferons/metabolism , Janus Kinase 1/deficiency , Job Syndrome/genetics , Mice , Mice, Knockout , Mutation , Phenotype , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , STAT1 Transcription Factor/deficiency , STAT2 Transcription Factor/deficiency , TYK2 Kinase/deficiency , TYK2 Kinase/genetics
13.
Cytotherapy ; 24(9): 884-891, 2022 09.
Article in English | MEDLINE | ID: mdl-35705447

ABSTRACT

Despite recent advances in the field of HSCT, viral infections remain a frequent causeof morbidity and mortality among HSCT recipients. Adoptive transfer of viral specific T cells has been successfully used both as prophylaxis and treatment of viral infections in immunocompromised HSCT recipients. Increasingly, precise risk stratification of HSCT recipients with infectious complications should incorporate not only pretransplant clinical criteria, but milestones of immune reconstitution as well. These factors can better identify those at highest risk of morbidity and mortality and identify a population of HSCT recipients in whom adoptive therapy with viral specific T cells should be considered for either prophylaxis or second line treatment early after inadequate response to first line antiviral therapy. Broadening these approaches to improve outcomes for transplant recipients in countries with limited resources is a major challenge. While the principles of risk stratification can be applied, early detection of viral reactivation as well as treatment is challenging in regions where commercial PCR assays and antiviral agents are not readily available.


Subject(s)
Hematopoietic Stem Cell Transplantation , Virus Diseases , Adoptive Transfer , Antiviral Agents/therapeutic use , Cell Engineering , Genetic Therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Virus Diseases/etiology , Virus Diseases/prevention & control
14.
BMC Infect Dis ; 22(1): 588, 2022 Jul 03.
Article in English | MEDLINE | ID: mdl-35786346

ABSTRACT

BACKGROUND: The associations between viral etiology of acute respiratory infections (ARI) with meteorological factors and air pollutants among children is not fully understood. This study aimed to explore the viral etiology among children hospitalized for ARI and the association of meteorological factors and air pollutants with children hospitalization due to viral ARI. METHODS: Electronic health record data about children (aged between 1 month and 14 years) admitted for ARI at Kiang Wu Hospital in Macao between 2014 and 2017 was analyzed retrospectively. xMAP multiplex assays were used to detect viruses in the nasopharyngeal swab and distributed-lag nonlinear model (DLNM) was used to evaluate associations. RESULTS: Among the 4880 cases of children hospitalization due to ARI, 3767 (77.2%) were tested positive for at least one virus and 676 (18%) exhibited multiple infections. Enterovirus (EV)/rhinovirus (HRV), adenovirus (ADV), respiratory syncytial virus (RSV) and influenza virus (IFV) were the most common viral pathogens associated with ARI and human bocavirus (hBOV) exhibited the highest multiple infection rates. Meteorological factors and air pollutants (PM10, PM2.5 and NO2) were associated with the risk of viral ARI hospitalization. The relative risk of viral infection increased with daily mean temperature but plateaued when temperature exceeded 23 °C, and increased when the relative humidity was < 70% and peaked at 50%. The effect of solar radiation was insignificant. Air pollutants (including PM10, PM2.5, NO2 and O3) showed strong and immediate effect on the incidence of viral infection. CONCLUSIONS: The effects of mean temperature, relative humidity and air pollutants should be taken into account when considering management of ARI among children.


Subject(s)
Air Pollutants , Respiratory Syncytial Virus, Human , Respiratory Tract Infections , Virus Diseases , Viruses , Air Pollutants/adverse effects , Child , Hospitalization , Humans , Infant , Macau , Meteorological Concepts , Nitrogen Dioxide , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/etiology , Retrospective Studies , Virus Diseases/epidemiology , Virus Diseases/etiology
15.
Int J Mol Sci ; 23(7)2022 Mar 24.
Article in English | MEDLINE | ID: mdl-35408912

ABSTRACT

Early post-transplant is the critical phase for the success of hematopoietic stem cell transplantation (HSCT). New viral infections and the reactivations associated with complete ablation of the recipient's T-cell immunity and inefficient reconstitution of the donor-derived system represent the main risks of HSCT. To date, the pharmacological treatments for post-HSCT viral infection-related complications have many limitations. Adoptive cell therapy (ACT) represents a new pharmacological strategy, allowing us to reconstitute the immune response to infectious agents in the post-HSC period. To demonstrate the potential advantage of this novel immunotherapy strategy, we report three cases of pediatric patients and the respective central nervous system complications after donor lymphocyte infusion.


Subject(s)
Communicable Diseases , Hematopoietic Stem Cell Transplantation , Neoplasms , Virus Diseases , Cell- and Tissue-Based Therapy/adverse effects , Child , Communicable Diseases/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunotherapy/adverse effects , Immunotherapy, Adoptive/adverse effects , Lymphocytes , Neoplasms/etiology , Virus Diseases/etiology , Virus Diseases/therapy
16.
Int J Mol Sci ; 23(1)2022 Jan 05.
Article in English | MEDLINE | ID: mdl-35009004

ABSTRACT

The nuclear lamina is the main component of the nuclear cytoskeleton that maintains the integrity of the nucleus. However, it represents a natural barrier for viruses replicating in the cell nucleus. The lamina blocks viruses from being trafficked to the nucleus for replication, but it also impedes the nuclear egress of the progeny of viral particles. Thus, viruses have evolved mechanisms to overcome this obstacle. Large viruses induce the assembly of multiprotein complexes that are anchored to the inner nuclear membrane. Important components of these complexes are the viral and cellular kinases phosphorylating the lamina and promoting its disaggregation, therefore allowing virus egress. Small viruses also use cellular kinases to induce lamina phosphorylation and the subsequent disruption in order to facilitate the import of viral particles during the early stages of infection or during their nuclear egress. Another component of the nuclear cytoskeleton, nuclear actin, is exploited by viruses for the intranuclear movement of their particles from the replication sites to the nuclear periphery. This study focuses on exploitation of the nuclear cytoskeleton by viruses, although this is just the beginning for many viruses, and promises to reveal the mechanisms and dynamic of physiological and pathological processes in the nucleus.


Subject(s)
Cell Nucleus/metabolism , Cytoskeleton/metabolism , Disease Susceptibility , Host-Pathogen Interactions , Virus Diseases/etiology , Virus Diseases/metabolism , Actins/metabolism , Animals , Cytoskeleton/genetics , Gene Expression Regulation, Viral , Humans , Lamins/metabolism , Nuclear Envelope/metabolism , Nuclear Lamina/metabolism , Species Specificity , Virus Replication
17.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055001

ABSTRACT

Non-coding RNAs, particularly lncRNAs and miRNAs, have recently been shown to regulate different steps in viral infections and induction of immune responses against viruses. Expressions of several host and viral lncRNAs have been found to be altered during viral infection. These lncRNAs can exert antiviral function via inhibition of viral infection or stimulation of antiviral immune response. Some other lncRNAs can promote viral replication or suppress antiviral responses. The current review summarizes the interaction between ncRNAs and herpes simplex virus, cytomegalovirus, and Epstein-Barr infections. The data presented in this review helps identify viral-related regulators and proposes novel strategies for the prevention and treatment of viral infection.


Subject(s)
Disease Susceptibility , Host-Pathogen Interactions/genetics , RNA, Untranslated , Virus Diseases/etiology , Virus Replication , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , DNA Viruses/physiology , Disease Models, Animal , Disease Susceptibility/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation, Viral/drug effects , Host-Pathogen Interactions/immunology , Humans , Molecular Diagnostic Techniques , Protein Binding , Species Specificity , Virus Diseases/diagnosis , Virus Diseases/drug therapy , Virus Diseases/metabolism , Virus Replication/drug effects
18.
Br J Haematol ; 194(1): 145-157, 2021 07.
Article in English | MEDLINE | ID: mdl-34124796

ABSTRACT

Community respiratory viral infections (CRVIs) are associated with pulmonary function impairment, alloimmune lung syndromes and inferior survival in human leucocyte antigen (HLA)-matched allogeneic haematopoietic stem cell transplant (HCT) recipients. Although the incidence of viral infections in HLA-haploidentical HCT recipients who receive post-transplant cyclophosphamide (PTCy)-based graft-versus-host disease (GVHD) prophylaxis is reportedly increased, there are insufficient data describing the incidence of CRVIs and the impact of donor source and PTCy on transplant outcomes. Analysing patients receiving their first HCT between 2012 and 2017 for acute myeloid leukaemia, acute lymphoblastic leukaemia and myelodysplastic syndromes, we describe comparative outcomes between matched sibling transplants receiving either calcineurin-based GVHD prophylaxis (SibCNI, N = 1605) or PTCy (SibCy, N = 403), and related haploidentical transplants receiving PTCy (HaploCy, N = 757). The incidence of CRVIs was higher for patients receiving PTCy, regardless of donor type. Patients in the HaploCy cohort who developed a CRVI by day +180 had both a higher risk of treatment-related mortality [hazard ratio (HR) 2⋅14, 99% confidence interval (CI) 1⋅13-4⋅07; P = 0⋅002] and inferior 2-year overall survival (HR 1⋅65, 99% CI 1⋅11-2⋅43; P = 0⋅001) compared to SibCNI with no CRVI. This finding justifies further research into long-term antiviral immune recovery, as well as development of preventive and treatment strategies to improve long-term outcomes in such patients.


Subject(s)
Community-Acquired Infections/etiology , Cyclophosphamide/adverse effects , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents/adverse effects , Respiratory Tract Infections/etiology , Transplantation, Haploidentical , Virus Diseases/etiology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Community-Acquired Infections/epidemiology , Cyclophosphamide/therapeutic use , Female , HLA Antigens/immunology , Humans , Immunosuppressive Agents/therapeutic use , Incidence , Kaplan-Meier Estimate , Leukemia/therapy , Living Donors , Male , Middle Aged , Myelodysplastic Syndromes/therapy , Proportional Hazards Models , Respiratory Tract Infections/epidemiology , Retrospective Studies , Siblings , Virus Diseases/epidemiology , Young Adult
19.
J Clin Immunol ; 41(4): 748-755, 2021 05.
Article in English | MEDLINE | ID: mdl-33462728

ABSTRACT

PURPOSE: Use of adoptive immunotherapy with virus-specific T cells (VST) in patients with inborn errors of immunity prior to hematopoietic stem cell transplantation (HSCT) has been reported in few patients. We report our experience, reviewing all the cases previously reported. METHODS: We report four children with inborn errors of immunity who received VST infusion in a pre-HSCT setting in two reference centers in Spain and review all inborn errors of immunity cases previously reported. RESULTS: Taking into account our four cases, nine children have been reported to receive VST prior to HSCT to date: 3 severe combined immunodeficiency, 2 CTPS1 deficiency, 1 dyskeratosis congenital, 1 ORAI1 deficiency, 1 Rothmund-Thomson syndrome, and 1 combined immunodeficiency without confirmed genetic defect. In four patients, immunotherapy resulted in clinical improvement, allowing to proceed to HSCT. In these cases, the infusion was started closely to viral diagnosis [mean time 28 days (IQR; 17-52 days)], and the VST was followed shortly thereafter by HSCT [mean time 28 days (IQR; 10-99 days)]. Viremia was controlled after HSCT in two cases (performed 7 and 36 days after the infusion). Multiple infusions were required in many cases. Five out of nine patients died before receiving HSCT. These patients presented with a prolonged and uncontrolled infection before VST administration [mean time from viral diagnosis to VST infusion was 176 days (IQR; 54-1687)]. CONCLUSIONS: In patients with inborn errors of immunity, the efficacy of VST for treating disseminated viral infections in pre-transplant settings seems to have a limited efficacy. However, this therapy could be used in a pre-emptive setting before severe viral disease occurs or closely to HSCT.


Subject(s)
Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Immune System Diseases/genetics , Immune System Diseases/therapy , Immunotherapy, Adoptive/methods , Preoperative Care , T-Lymphocytes/immunology , Disease Management , Disease Susceptibility , Genetic Diseases, Inborn/complications , Genetic Diseases, Inborn/diagnosis , Hematopoietic Stem Cell Transplantation/methods , Humans , Immune System Diseases/complications , Immune System Diseases/diagnosis , Immunotherapy, Adoptive/adverse effects , Preoperative Care/methods , T-Cell Antigen Receptor Specificity , T-Lymphocytes/metabolism , Treatment Failure , Treatment Outcome , Virus Diseases/etiology
20.
Crit Rev Microbiol ; 47(3): 307-322, 2021 May.
Article in English | MEDLINE | ID: mdl-33570448

ABSTRACT

The ongoing COVID-19 pandemic has made us wonder what led to its occurrence and what can be done to avoid such events in the future. As we document, one changing circumstance that is resulting in the emergence and changing the expression of viral diseases in both plants and animals is climate change. Of note, the rapidly changing environment and weather conditions such as excessive flooding, droughts, and forest fires have raised concerns about the global ecosystem's security, sustainability, and balance. In this review, we discuss the main consequences of climate change and link these to how they impact the appearance of new viral pathogens, how they may facilitate transmission between usual and novel hosts, and how they may also affect the host's ability to manage the infection. We emphasize how changes in temperature and humidity and other events associated with climate change influence the reservoirs of viral infections, their transmission by insects and other intermediates, their survival outside the host as well the success of infection in plants and animals. We conclude that climate change has mainly detrimental consequences for the emergence, transmission, and outcome of viral infections and plead the case for halting and hopefully reversing this dangerous event.


Subject(s)
COVID-19/transmission , Climate Change , Communicable Diseases, Emerging/transmission , Plant Diseases/virology , Virus Diseases/transmission , Animals , Aquatic Organisms/virology , COVID-19/complications , COVID-19/etiology , COVID-19/immunology , Chiroptera/virology , Communicable Diseases, Emerging/complications , Communicable Diseases, Emerging/etiology , Communicable Diseases, Emerging/immunology , Crops, Agricultural/virology , Disease Reservoirs/virology , Disease Vectors/classification , Food Supply , Humans , Humidity , Plant Diseases/immunology , Primate Diseases/transmission , Primate Diseases/virology , Primates , Rain , Seasons , Temperature , Virus Diseases/complications , Virus Diseases/etiology , Virus Diseases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL